IL259964B - Adeno-associated viral vectors for treating mucolipidosis type ii - Google Patents

Adeno-associated viral vectors for treating mucolipidosis type ii

Info

Publication number
IL259964B
IL259964B IL259964A IL25996418A IL259964B IL 259964 B IL259964 B IL 259964B IL 259964 A IL259964 A IL 259964A IL 25996418 A IL25996418 A IL 25996418A IL 259964 B IL259964 B IL 259964B
Authority
IL
Israel
Prior art keywords
aav
raav
gnptab
optionally
nucleic acid
Prior art date
Application number
IL259964A
Other languages
Hebrew (he)
Other versions
IL259964A (en
Original Assignee
Genzyme Corp
Samsung Life Public Welfare Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genzyme Corp, Samsung Life Public Welfare Foundation filed Critical Genzyme Corp
Publication of IL259964A publication Critical patent/IL259964A/en
Publication of IL259964B publication Critical patent/IL259964B/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knockout animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0075Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0083Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the administration regime
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • A61K49/0008Screening agents using (non-human) animal models or transgenic animal models or chimeric hosts, e.g. Alzheimer disease animal model, transgenic model for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/10Laxatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/16Central respiratory analeptics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1288Transferases for other substituted phosphate groups (2.7.8)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/08Transferases for other substituted phosphate groups (2.7.8)
    • C12Y207/08017UDP-N-acetylglucosamine--lysosomal-enzyme N-acetylglucosaminephosphotransferase (2.7.8.17)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • A01K2217/077Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out heterozygous knock out animals displaying phenotype
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0306Animal model for genetic diseases
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14132Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14142Use of virus, viral particle or viral elements as a vector virus or viral particle as vehicle, e.g. encapsulating small organic molecule
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14151Methods of production or purification of viral material
    • C12N2750/14152Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/15Vector systems having a special element relevant for transcription chimeric enhancer/promoter combination
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/50Vector systems having a special element relevant for transcription regulating RNA stability, not being an intron, e.g. poly A signal

Description

259964/3 ADENO-ASSOCIATED VIRAL VECTORS FOR TREATING MUCOLIPIDOSIS TYPE II id="p-1" id="p-1" id="p-1" id="p-1" id="p-1"
[0001] id="p-2" id="p-2" id="p-2" id="p-2" id="p-2"
[0002] FIELD OF THE INVENTION id="p-3" id="p-3" id="p-3" id="p-3" id="p-3"
[0003] The present invention relates to rAAV vectors, particles, compositions, for treating mucolipidosis type II and/or type III, as well as methods, kits, and uses related thereto.
BRIEF SUMMARY OF THE INVENTION id="p-4" id="p-4" id="p-4" id="p-4" id="p-4"
[0004] Mucolipidosis types II and III (ML II; ML III) are autosomal recessive lysosomal storage diseases characterized by a deficiency of UDP-GlcNAc;Lysosomal enzyme N- acetylglucosamine-1 -phosphotransferase (abbreviated GlcNAc-1 -phosphotransferase). GlcNAc-1-phosphotransferase is a hexameric enzyme complex consisting of three different subunits: a2, P2, and y2. The a2, and P2 subunits contain the catalytic activity and are encoded by a single gene, GNPTAB. Mutations in GNPTAB that result in a complete loss of enzymatic activity are found in patients with MLII. Mucolipidosis II is highly progressive, and patients rarely survive past the first decade of life. In addition, the available treatment options for ML II remain limited. Only bone marrow transplantation has been attempted in a small number of patients. In contrast, mutations in GNPTAB resulting in a partial reduction in GlcNAc-1- phosphotransferase activity are found in patients with MLIII. These patients generally show 259964/3 less severe symptoms and slower disease progression but still present with serious health problems such as skeletal abnormalities, developmental delay, and cardiomegaly. id="p-5" id="p-5" id="p-5" id="p-5" id="p-5"
[0005] The availability of AAV serotypes that display tissue-specific tropism and promote sustained expression of transgenes offers the possibility of AAV-mediated gene therapy for the systemic treatment of lysosomal disease, including ML II and ML III. Accordingly, investigation of AAV-mediated treatment of ML II/DI is important to establish whether this approach represents a potential therapeutic strategy for this devastating disease. However, before AAV-mediated treatment of MLII/III can be examined, technical limitations need to be overcome. The coding sequence of GNPTAB (over 5.6 kb in humans) is longer than the endogenous AAV genome (approximately 4.7 kb). Therefore, AAV vectors that can accommodate GNPTAB along with functional promoter/enhancer sequences and the other components of the AAV genome while still facilitating efficient packaging into viral particles are highly advantageous. id="p-6" id="p-6" id="p-6" id="p-6" id="p-6"
[0006] The invention provides s recombinant adeno-associated virus (rAAV) vector comprising nucleic acid encoding N-acetylglucosamine-1-phosphate transferase (GNPTAB) and at least one AAV inverted terminal repeat (ITR). In some embodiments, the GNPTAB comprises the alpha and beta subunits. In some embodiments, the GNPTAB is operably linked to a promoter. In some embodiments, the GNPTAB is a human GNPTAB. In some embodiments, the GNPTAB comprises an amino acid sequence that is at least about 80%, at least about 85%, at least about 90% or at least about 95% identical to the amino acid sequence of SEQ ID NO:1. In some embodiments, the GNPTAB comprises the amino acid sequence of SEQ ID NO:1. In some embodiments, the promoter is a CMV enhancer/chicken beta-actin (CBA) promoter. In some embodiments, the CBA promoter is a modified CBA promoter. In some embodiments, the modified CBA promoter is a truncated CBA promoter. In some embodiments, the CMV enhancer is a shortened CMV enhancer. In some embodiments, the vector comprises an intron. In some embodiments, the intron is an MVM intron. In some embodiments, the vector comprises a polyadenylation sequence. In some embodiments, the polyadenylation sequence is a bovine growth hormone polyadenylation sequence. In some embodiments, the AAV terminal repeat is an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAVrh8R, AAV9, AAV10, AAVrh10, AAV11, AAV12, 259964/3 AAV2R471A, AAV DJ, a goat AAV, bovine AAV, or mouse AAV serotype ITR. In some embodiments, the rAAV vector comprises two ITRs. id="p-7" id="p-7" id="p-7" id="p-7" id="p-7"
[0007] In some aspects, the invention provides a rAAV particle comprising the rAAV vector of any one of the above embodiments. In some embodiments, the AAV particle comprises an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAVrh8R, AAV9, AAV10, AAVrh10, AAV11, AAV12, AAV2R471A, AAV2/2-7m8, AAV DJ, AAV2 N587A, AAV2 E548A, AAV2 N708A, AAV V708K, goat AAV, AAV1/AAV2 chimeric, bovine AAV, mouse AAV, or rAAV2/HBoV1 serotype capsid. In some embodiments, the rAAV particle comprises one or more ITRs and capsid derived from the same AAV serotype. In some embodiments, the rAAV particle comprises one or more ITRs derived from a different AAV serotype than capsid of the rAAV viral particles. In some embodiments, the rAAV particle comprises an AAV8 capsid, and wherein the vector comprises AAV2 ITRs. In some embodiments, the rAAV particle is produced by transfecting a host cell with nucleic acid encoding the rAAV vector and nucleic acid encoding AAV rep and cap functions, and providing nucleic acid encoding AAV helper functions. In some embodiments, the AAV helper functions are provided by transfecting the host cell with nucleic acid encoding the AAV helper functions. In some embodiments, the AAV helper functions are provided by infecting the host cell with an AAV helper virus that provides the AAV helper functions. In some embodiments, the AAV helper virus is an adenovirus, a herpes simplex virus or a baculovirus. In some embodiments, the rAAV particle is produced by an AAV producer cell comprising nucleic acid encoding the rAAV vector and nucleic acid encoding AAV rep and cap functions, and providing nucleic acid encoding AAV helper functions. In some embodiments, the AAV producer cell comprises nucleic acid encoding AAV helper functions. In some embodiments, the AAV helper functions are provided by infecting the AAV producer cells with an AAV helper virus that provides the AAV helper functions. In some embodiments, the AAV helper virus is an adenovirus, a herpes simplex virus, or a baculovirus. In some embodiments, the invention provides a pharmaceutical composition comprising any of the rAAV particles described herein. id="p-8" id="p-8" id="p-8" id="p-8" id="p-8"
[0008] In some aspects, the invention provides a method for treating mucolipidosis type II (ML II) or mucolipidosis type III (ML III) in a mammal comprising administering to the 259964/3 mammal an effective amount of any of the rAAV particle described herein or the pharmaceutical composition described herein. id="p-9" id="p-9" id="p-9" id="p-9" id="p-9"
[0009] In some aspects, the invention provides a method for treating mucolipidosis type II (ML II) or mucolipidosis type III (ML III) in a mammal comprising administering to the mammal an effective amount of rAAV particles wherein the rAAV particles comprise a rAAV vector, wherein the rAAV vector comprises nucleic acid encoding N-acetylglucosamine-1- phosphate transferase (GNPTAB) and at least one AAV ITR. In some embodiments, the invention provides a method of maintaining or increasing body size in a mammal with mucolipidosis type II (ML II) or mucolipidosis type III (ML III) comprising administering to the mammal an effective amount of rAAV particles wherein the rAAV particles comprise a rAAV vector, wherein the rAAV vector comprises nucleic acid encoding N- acetylglucosamine-1-phosphate transferase (GNPTAB) and at least one AAV ITR, wherein expression of the GNPTAB results in maintenance of or an increase in body weight gain and/or maintenance of or an increase in body height gain. In some embodiments, the invention provides a method of preventing the reduction of body size in a mammal with mucolipidosis type II (ML II) or mucolipidosis type III (ML III) comprising administering to the mammal an effective amount of rAAV particles wherein the rAAV particles comprise a rAAV vector, wherein the rAAV vector comprises nucleic acid encoding N-acetylglucosamine-1-phosphate transferase (GNPTAB) and at least one AAV ITR, wherein expression of the GNPTAB prevents the reduction of body weight. In some embodiments, the invention provides a method of maintaining or increasing bone mineral content in a mammal with mucolipidosis type II (ML II) or mucolipidosis type III (ML III) comprising administering to the mammal an effective amount of rAAV particles wherein the rAAV particles comprise a rAAV vector, wherein the rAAV vector comprises nucleic acid encoding GNPTAB and at least one AAV ITR, wherein expression of the GNPTAB results in maintenance or an increase in bone mineral content. In some embodiments, the invention provides a method of preventing the reduction of bone mineral content in a mammal with mucolipidosis type II (ML II) or mucolipidosis type III (ML III) comprising administering to the mammal an effective amount of rAAV particles wherein the rAAV particles comprise a rAAV vector, wherein the rAAV vector comprises nucleic acid encoding GNPTAB and at least one AAV ITR, wherein expression of the GNPTAB prevents reduction in bone mineral content. In some embodiments, the invention 259964/3 provides a method of maintaining or increasing bone mineral density in a mammal with mucolipidosis type II (ML II) or mucolipidosis type III (ML III) comprising administering to the mammal an effective amount of rAAV particles wherein the rAAV particles comprise a rAAV vector, wherein the rAAV vector comprises nucleic acid encoding GNPTAB and at least one AAV ITR, wherein expression of the GNPTAB results in maintenance or an increase in bone mineral density. In some embodiments, the invention provides a method of preventing the reduction of bone mineral density in a mammal with mucolipidosis type II (ML II) or mucolipidosis type III (ML III) comprising administering to the mammal an effective amount of rAAV particles wherein the rAAV particles comprise a rAAV vector, wherein the rAAV vector comprises nucleic acid encoding GNPTAB and at least one AAV ITR, wherein expression of the GNPTAB prevents reduction in bone mineral density. id="p-10" id="p-10" id="p-10" id="p-10" id="p-10"
[0010] In some embodiments of the above methods, the treatment ameliorates one or more symptoms of ML II or ML III, wherein the one or more symptoms of ML II or ML III are skeletal defects, cognitive deficits, delays in the development of gross and fine motor skills, hearing loss, lack of muscle tone, protruded abdomen, umbilical hernias, progressive mucosal thickening of the airways, frequent respiratory infections, thickening and insufficiency of the mitral valve, constipation or diarrhea. In some embodiments, the treatment delays to progression of one or more symptoms of ML II or ML III, wherein the one or more symptoms of ML II or ML III are skeletal defects, cognitive deficits, delays in the development of gross and fine motor skills, hearing loss, lack of muscle tone, protruded abdomen, umbilical hernias, progressive mucosal thickening of the airways, frequent respiratory infections, thickening and insufficiency of the mitral valve, constipation or diarrhea. In some embodiments, the invention provides a method of ameliorating one or more symptoms of ML II or ML III in a mammal comprising administering to the mammal an effective amount of rAAV particles wherein the rAAV particles comprise a rAAV vector, wherein the rAAV vector comprises nucleic acid encoding GNPTAB and at least one AAV ITR; wherein the one or more symptoms of ML II or ML III are skeletal defects, cognitive deficits, delays in the development of gross and fine motor skills, hearing loss, lack of muscle tone, protruded abdomen, umbilical hernias, progressive mucosal thickening of the airways, frequent respiratory infections, thickening and insufficiency of the mitral valve, constipation or diarrhea. In some embodiments, the invention provides a method of delaying the progression of one or more symptoms of ML II or ML III in 259964/3 a mammal comprising administering to the mammal an effective amount of rAAV particles wherein the rAAV particles comprise a rAAV vector, wherein the rAAV vector comprises nucleic acid encoding GNPTAB and at least one AAV ITR; wherein the one or more symptoms of ML II or ML III are skeletal defects, cognitive deficits, delays in the development of gross and fine motor skills, hearing loss, lack of muscle tone, protruded abdomen, umbilical hernias, progressive mucosal thickening of the airways, frequent respiratory infections, thickening and insufficiency of the mitral valve, constipation or diarrhea. id="p-11" id="p-11" id="p-11" id="p-11" id="p-11"
[0011] In some embodiments of the above methods, the GNPTAB is operably linked to a promoter. In some embodiments, the GNPTAB is a human GNPTAB. In some embodiments, the GNPTAB comprises an amino acid sequence that is at least about 80% identical to the amino acid sequence of SEQ ID NO:1. In some embodiments, the GNPTAB comprises the amino acid sequence of SEQ ID NO:1. In some embodiments, the promoter is a CMV enhancer/chicken beta-actin (CBA) promoter. In some embodiments, the CBA promoter is a modified CBA promoter. In some embodiments, the modified CBA promoter is a truncated CBA promoter. In some embodiments, the CMV enhancer is a shortened CMV enhancer. In some embodiments, the vector comprises an intron. In some embodiments, the intron is an MVM intron. In some embodiments, the vector comprises a polyadenylation sequence. In some embodiments, the polyadenylation sequence is a bovine growth hormone polyadenylation sequence. In some embodiments, the AAV terminal repeat is an AAV1, AAV2, AAV3,AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAVrh8R, AAV9, AAV10, AAVrh10, AAV11, AAV12, AAV2R471A, AAV DJ, a goat AAV, bovine AAV, or mouse AAV serotype ITR. In some embodiments, the rAAV vector comprises two ITRs. In some embodiments, the AAV particle comprises an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAVrh8R, AAV9, AAV10, AAVrh10, AAV11, AAV12, AAV2R471A, AAV2/2- 7m8, AAV DJ, AAV2 N587A, AAV2 E548A, AAV2 N708A, AAV V708K, goat AAV, AAV1/AAV2 chimeric, bovine AAV, mouse AAV, or rAAV2/HBoV1 serotype capsid. In some embodiments, the rAAV particle comprises one or more ITRs and capsid derived from the same AAV serotype. In some embodiments, the rAAV particle comprises one or more ITRs derived from a different AAV serotype than capsid of the rAAV viral particles. In some embodiments, the rAAV particle comprises an AAV8 capsid, and wherein the vector comprises AAV2 ITRs. 259964/3 id="p-12" id="p-12" id="p-12" id="p-12" id="p-12"
[0012] In some embodiments of the above embodiments, the rAAV particle is produced by transfecting a host cell with nucleic acid encoding the rAAV vector and nucleic acid encoding AAV rep and cap functions, and providing nucleic acid encoding AAV helper functions. In some embodiments, the AAV helper functions are provided by transfecting the host cell with nucleic acid encoding the AAV helper functions. In some embodiments, the AAV helper functions are provided by infecting the host cell with an AAV helper virus that provides the AAV helper functions. In some embodiments, the AAV helper virus is an adenovirus, a herpes simplex virus, or a baculovirus. In some embodiments, the rAAV particle is produced by an AAV producer cell comprising nucleic acid encoding the rAAV vector and nucleic acid encoding AAV rep and cap functions, and providing nucleic acid encoding AAV helper functions. In some embodiments, the AAV producer cell comprises nucleic acid encoding AAV helper functions. In some embodiments, the AAV helper functions are provided by infecting the AAV producer cells with an AAV helper virus that provides the AAV helper functions. In some embodiments, the AAV helper virus is an adenovirus, a herpes simplex virus, or a baculovirus. id="p-13" id="p-13" id="p-13" id="p-13" id="p-13"
[0013] In some embodiments of the above methods, the mammal is a human. In some embodiments, the human is a pediatric subject. In some embodiments, the human is a young adult. id="p-14" id="p-14" id="p-14" id="p-14" id="p-14"
[0014] In some embodiments of the above methods, the rAAV is administered intravenously, intraperitoneally, intra-arterially, intramuscularly, subcutaneously, or intrahepatically. In some embodiments, the rAAV is administered intravenously. In some embodiments, the rAAV is administered to more than one location. In some embodiments, the administration is repeated. In some embodiments, the rAAV viral particles are in a pharmaceutical composition. In some embodiments, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier. id="p-15" id="p-15" id="p-15" id="p-15" id="p-15"
[0015] In some aspects, the invention provides the use of any pharmaceutical composition as described herein the manufacture of a medicament for treating ML II or ML III in a mammal. In some embodiments, the invention provides the use of any pharmaceutical composition as described herein in the manufacture of a medicament for use in any of the methods described 259964/3 herein. In some embodiments, the invention provides the use of any of the rAAV particles as described herein in the manufacture of a medicament for treating ML II or ML III in a mammal. In some embodiments, the invention provides the use of any of the rAAV particles described herein the manufacture of a medicament for use in any of the methods as described herein. In some embodiments, the invention provides the use of any of the pharmaceutical compositions described herein for treating ML II or ML III in a mammal. In some embodiments, the invention provides the use of any of the pharmaceutical compositions described herein for use in any of the methods described herein. In some embodiments, the invention provides the use of any of the recombinant AAV described herein for treating ML II or ML III in a mammal. In some embodiments, the invention provides the use of any of the recombinant AAV described herein for use in any of the methods described herein. In some embodiments, the invention provides the use any of the pharmaceutical compositions described herein in the manufacture of a medicament for ameliorating one or more symptoms of ML II or ML III in a mammal or delaying the progression of one or more symptoms of ML II or ML III in a mammal. In some embodiments, the invention provides the use of any rAAV particles described herein in the manufacture of a medicament for ameliorating one or more symptoms of ML II or ML III in a mammal or delaying the progression of one or more symptoms of MLII or ML III in a mammal. In some embodiments, the invention provides the use of any pharmaceutical composition described herein for ameliorating one or more symptoms of ML II or ML III in a mammal or delaying the progression of one or more symptoms of ML II or MLIII in a mammal. In some embodiments, the invention provides the use of any recombinant AAV described herein for ameliorating one or more symptoms of ML II or ML III in a mammal or delaying the progression of one or more symptoms of ML II or ML III in a mammal. In some embodiments, the one or more symptoms of ML II or ML III are skeletal defects, cognitive deficits, delays in the development of gross and fine motor skills, hearing loss, lack of muscle tone, protruded abdomen, umbilical hernias, progressive mucosal thickening of the airways, frequent respiratory infections, thickening and insufficiency of the mitral valve, constipation or diarrhea. In some embodiments, the mammal is a human. id="p-16" id="p-16" id="p-16" id="p-16" id="p-16"
[0016] In some aspects, the invention provides a kit comprising any of the rAAV vectors described herein, any of the rAAV particles described herein or any pharmaceutical composition as described herein. In some embodiments, the kit is for treating ML II or ML III 259964/3 according any of the methods as described herein. In some embodiments, the kit comprises any of the rAAV vectors as described herein, any of the rAAV particles as described herein or any pharmaceutical composition as described herein. In some embodiments, the kit further comprises one or more buffers or pharmaceutically acceptable excipients. In some embodiments, the kit further comprises instructions for use in treating ML II and/or ML III. id="p-17" id="p-17" id="p-17" id="p-17" id="p-17"
[0017] In some aspects, the invention provides an animal model of Mucoliposis II (ML II) wherein at least one allele of a N-acetylglucosamine-1-phosphate transferase (GNPTAB) gene comprises a deletion located between exons 12 and exon 20. In some embodiments, at least one allele of the GNPTAB gene comprises a deletion spanning exons 12 and exon 20. In some embodiments, the animal is homozygous for the deletion in the GNPTAB gene. In some embodiments, the animal is heterozygous for the deletion in the GNPTAB gene. In some embodiments, a portion of the GNPTAB gene is replaced by a gene encoding a reporter and/or selectable marker. In some embodiments, the selectable marker confers resistance to neomycin. In some embodiments, the animal is a mammal. In some embodiments, the mammal is a rodent. In some embodiments, the rodent is a mouse. In some embodiments, the mouse has a genetic background derived from 129/Sv and/or C57Bl/6. In some embodiments, the animal is immunocompetent or immunodeficient. id="p-18" id="p-18" id="p-18" id="p-18" id="p-18"
[0018] In some aspects, the invention provides a method of generating an animal model of Mucolipidosis II (ML II) comprising introducing a deletion between exons 12 and 20 in at least one allele of the GNPTAB gene on the animal. In some embodiments, at least one allele of the GNPTAB gene comprises a deletion spanning exons 12 and exon 20. In some embodiments, the animal is bred to be homozygous for the deletion in the GNPTAB gene. In some embodiments, the animal is bred to be heterozygous for the deletion in the GNPTAB gene. In some embodiments, a portion of the GNPTAB gene is replaced by a gene encoding a reporter and/or selectable marker. In some embodiments, the selectable marker confers resistance to neomycin. In some embodiments, the animal is a mammal. In some embodiments, the mammal is a rodent. In some embodiments, the rodent is a mouse. In some embodiments, the mouse has a genetic background derived from 129/Sv and/or C57Bl/6. In some embodiments, the animal is immunocompetent or immunodeficient. 259964/3 id="p-19" id="p-19" id="p-19" id="p-19" id="p-19"
[0019] In some aspects, the invention provides a method for evaluating an agent for treatment of Mucolipidosis II (ML II) comprising administering the agent to the animal model as described herein, wherein amelioration one or more symptoms of ML II indicates the agent may provide beneficial treatment of ML II. In some embodiments, the symptom of ML II is decreased body weight, decreased bone density, decreased bone mineral content, skeletal defects, cognitive deficits, delays in the development of gross and fine motor skills, hearing loss, lack of muscle tone, protruded abdomen, umbilical hernias, progressive mucosal thickening of the airways, frequent respiratory infections, thickening and insufficiency of the mitral valve, constipation and/or diarrhea. In some embodiments, the agent is a small molecule, a polypeptide, an antibody, a nucleic acid or a recombinant viral particle. id="p-20" id="p-20" id="p-20" id="p-20" id="p-20"
[0020] BRIEF DESCRIPTION OF THE DRAWINGS id="p-21" id="p-21" id="p-21" id="p-21" id="p-21"
[0021] FIG. 1A provides a diagram of the mouse GNPTAB gene structure and the genomic insertion site of the gene trapping vector used for generating GNPTAB knockout mice. id="p-22" id="p-22" id="p-22" id="p-22" id="p-22"
[0022] FIG. 1B is a Southern blot showing mouse ES clones used for generating GNPTAB knockout mice. id="p-23" id="p-23" id="p-23" id="p-23" id="p-23"
[0023] FIGS. 2A-2C show the growth retardation present in GNPTAB knock-out mice. (FIG. 2A) Body weight (g) of wild type (+/+), heterozygous (+/-), and homozygous (-/-) mice at 6 weeks of age (***; p<0.000l ; Bonterroni multiple comparison test). (FIG. 2B) Naso-anal length (mm) of wild type, heterozygous, and homozygous mice at 6 weeks of (*; p<0.02 ; Bonterroni multiple comparison test). (FIG. 2C) Gross morphology of wild type and homozygous mice. id="p-24" id="p-24" id="p-24" id="p-24" id="p-24"
[0024] FIGS. 3A & 3B show light microscopic images of representative sections of hematoxylin and eosin-stained femoral cartilage from wild type (FIG. 3A) and homozygous knockout (FIG. 3B) mice. 259964/3 id="p-25" id="p-25" id="p-25" id="p-25" id="p-25"
[0025] FIGs. 4A-4F demonstrates the accumulation of autolysosomes in the salivary glands of knockout (KO) mice. (FIGS. 4A-C) EM showing an overview of wild type mouse salivary gland acinus, composed of mucous and serous cells. (FIG. 4D) Overview of a KO salivary gland acinus. The overall architecture is disrupted by the massive accumulation of large vacuoles in KO. (FIGS. 4E-F) Higher magnification of a vacuole from (FIG. 4D), which is surrounded by a single membrane and contains un-degraded material. Area of magnification is indicated by the box in (FIG. 4D). AL, autolysosome; Mu, mucous cell; N, nucleus; SG; secretory granule. id="p-26" id="p-26" id="p-26" id="p-26" id="p-26"
[0026] FIGS. 5A-5D show lysosomal enzyme activities in sera of wild type (open circles) and KO mice (filled circles), as labeled. Activity of N-acetylglucosaminidase (FIG. 5A), P- hexosaminidase A (FIG. 5B), P־galactosidase (FIG. 5C), and P־glucuronidase (FIG. 5D) are shown. id="p-27" id="p-27" id="p-27" id="p-27" id="p-27"
[0027] FIGS. 6A & 6B provide an overview of the experimental timeline for injections. (FIG. 6A) Timelines for long term treatment of the study for mice injected with viral vector in weeks-old age. (FIG. 6B) The total number of mice (n) injected for each treatment group is indicated. id="p-28" id="p-28" id="p-28" id="p-28" id="p-28"
[0028] FIG. 7A depicts a schematic of the pAAV2/8־GNPTAB vector containing mouse GNPTAB cDNA. The mouse GNPTAB cDNA sequence is based on GenBank accession number NM_001004164.2. The nucleotide sequence was codon-optimized for expression in mouse. The amino acid sequence is unchanged. id="p-29" id="p-29" id="p-29" id="p-29" id="p-29"
[0029] FIG. 7B shows the quantitative analysis of livers from KO mice injected with AAV- GNPTAB as compared to their control littermates. id="p-30" id="p-30" id="p-30" id="p-30" id="p-30"
[0030] FIGS. 8A & 8B show the body weight change from starting weight over time for control and AAV-GNPTAB treated KO mice. (FIG. 8A) Total body weight of control mice and AAV- GNPTAB treated KO mice (*p< 0.05 Dunnett multiple comparison test). (FIG. 8B) Data expressed as amount of weight change from starting weight over time. id="p-31" id="p-31" id="p-31" id="p-31" id="p-31"
[0031] FIGS. 9A & 9B show the height change from starting height over time for control and AAV-GNPTAB treated KO mice. (FIG. 9A) Data expressed as the ratio of body length 259964/3 compared before and 6 weeks post injection (FIG. 9B) Data expressed as the ratio of body length compared before and 32 weeks post injection. id="p-32" id="p-32" id="p-32" id="p-32" id="p-32"
[0032] FIGS. 10A-10C provide histograms showing levels of bone mineral density before injection (FIG. 10A), at 16 weeks post injection (FIG. 10B), and at 32 weeks post injection (FIG. 10C). (FIG. 10A) The data from homozygous and homozygous treated with AAV- GNPTAB were compared with that of wild type and heterozygous. (f;p<0.05, {; p<0.compared with wild type, *;p<0.02, **; p<0.002 compared with heterozygous). AAV- GNPTAB treatment resulted in a statistically significant increase in BMD ratio (post/before Tx) in homozygous mice post16 weeks treatment (FIG. 10B) and 32 weeks treatment (FIG. 10C). (FIG. 10B) 16 weeks after treatment, GNPTAB null mice treated with AAV-GNPTAB showed a significantly increase in BMD ratio than other mice (**; P<0.02) (FIG. 10C) weeks after treatment, significant differences in BMD ratio was observed in GNPTAB treated mice. (#; p<0.05, **; p<0.02). P values were determined by two-tailed, unpaired t-test analyses. Data are presented as Mean ± SEM. id="p-33" id="p-33" id="p-33" id="p-33" id="p-33"
[0033] FIGS. 11A-11C provide histograms provide histograms showing levels of bone mineral content before injection (FIG. 11A), at 16 weeks post injection (FIG. 11B), and at weeks post injection (FIG. 11C). (FIG. 11A) The data from homozygous, and homozygous treated with AAV-GNPTAB were compared with that of wild type and heterozygous. (f; p<0.05, {;p<0.02 compared with wild type, *;p<0.02, **;p<0.002 compared with heterozygous). AAV-GNPTAB treatment resulted in a statistically significant increase in BMD ratio (post/before Tx) in homozygous mice post16 weeks treatment (FIG. 11B) and 32 weeks treatment (FIG. 11C). (FIG. 11B) 16 weeks after treatment, GNPTAB null mice treated with AAV-GNPTAB showed a significantly increase in BMD ratio than other mice (**; P<0.02) (FIG. 11C) 32 weeks after treatment, significant differences in BMD ratio was observed in GNPTAB treated mice. (#; p<0.05, **; p<0.02). P values were determined by two-tailed, unpaired t-test analyses. Data are presented as Mean ± SEM. id="p-34" id="p-34" id="p-34" id="p-34" id="p-34"
[0034] FIGS. 12A & 12B provide histograms showing levels of percent lean mass before injection (FIG. 12A) and change of percent lean mass 32 weeks post injection (FIG. 12B). (FIG. 12A) The data from homozygous, and homozygous treated with AAV-GNPTAB were 259964/3 compared with that of wild type and heterozygous. (f; p<0.02 compared with wild type, *; p<0.05, **; p<0.001 compared with heterozygous). (FIG. 12B) 32 weeks after treatment, no change in % lean mass was observed in AAV-GNPTAB treated mice. P values were determined by two-tailed, unpaired t-test analyses. Data are presented as Mean ± SEM.
DETAILED DESCRIPTION id="p-35" id="p-35" id="p-35" id="p-35" id="p-35"
[0035] In some aspects, the invention provides a recombinant adeno-associated virus (rAAV) vector comprising nucleic acid encoding N-acetylglucosamine-1-phosphate transferase, alpha and beta subunits (GNPTAB) and at least one AAV inverted terminal repeat (ITR). Further provided herein are rAAV particles comprising a rAAV vector of the present disclosure, as well as pharmaceutical compositions comprising a rAAV particle of the present disclosure. id="p-36" id="p-36" id="p-36" id="p-36" id="p-36"
[0036] In some aspects, the invention further provides methods for treating mucolipidosis type II (ML II) or mucolipidosis type III (ML III) in a mammal comprising administering to the mammal an effective amount of rAAV particles, where the rAAV particles comprise a rAAV vector, and the rAAV vector comprises nucleic acid encoding GNPTAB and at least one AAV ITR. Further provided herein are methods for increasing body size, bone mineral content, and/or bone mineral density in a mammal with mucolipidosis type II (ML II) or mucolipidosis type III (ML III) comprising administering to the mammal an effective amount of rAAV particles, where the rAAV particles comprise a rAAV vector, and the rAAV vector comprises nucleic acid encoding GNPTAB and at least one AAV ITR. In some embodiments, the expression of the GNPTAB results in an increase in body size, bone mineral content, and/or bone mineral density. id="p-37" id="p-37" id="p-37" id="p-37" id="p-37"
[0037] In some aspects, the invention yet further provides uses and/or kits for treating ML II or ML III, e.g., using a rAAV vector, rAAV particle, or pharmaceutical composition of the present disclosure. 259964/3 I. General Techniques id="p-38" id="p-38" id="p-38" id="p-38" id="p-38"
[0038] The techniques and procedures described or referenced herein are generally well understood and commonly employed using conventional methodology by those skilled in the art, such as, for example, the widely utilized methodologies described in Molecular Cloning: A Laboratory Manual (Sambrook et al., 4th ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 2012); Current Protocols in Molecular Biology (F.M. Ausubel, et al. eds., 2003); the series Methods in Enzymology (Academic Press, Inc.); PCR 2: A Practical Approach (M.J. MacPherson, B.D. Hames and G.R. Taylor eds., 1995); Antibodies, A Laboratory Manual (Harlow and Lane, eds., 1988); Culture of Animal Cells: A Manual of Basic Technique and Specialized Applications (R.I. Freshney, 6th ed., J. Wiley and Sons,2010); Oligonucleotide Synthesis (M.J. Gait, ed., 1984); Methods in Molecular Biology, Humana Press; Cell Biology: A Laboratory Notebook (J.E. Cellis, ed., Academic Press, 1998); Introduction to Cell and Tissue Culture (J.P. Mather and P.E. Roberts, Plenum Press, 1998); Cell and Tissue Culture: Laboratory Procedures (A. Doyle, J.B. Griffiths, and D.G. Newell, eds., J. Wiley and Sons, 1993-8); Handbook of Experimental Immunology (D.M. Weir and C.C. Blackwell, eds., 1996); Gene Transfer Vectors for Mammalian Cells (J.M. Miller and M.P. Calos, eds., 1987); PCR: The Polymerase Chain Reaction, (Mullis et al., eds., 1994); Current Protocols in Immunology (J.E. Coligan et al., eds., 1991); Short Protocols in Molecular Biology (Ausubel et al., eds., J. Wiley and Sons, 2002); Immunobiology (C.A. Janeway et al., 2004); Antibodies (P. Finch, 1997); Antibodies: A Practical Approach (D. Catty., ed., IRL Press, 1988-1989); Monoclonal Antibodies: A Practical Approach (P.Shepherd and C. Dean, eds., Oxford University Press, 2000); Using Antibodies: A Laboratory Manual (E. Harlow and D. Lane, Cold Spring Harbor Laboratory Press, 1999); The Antibodies (M. Zanetti and J. D. Capra, eds., Harwood Academic Publishers, 1995); and Cancer: Principles and Practice of Oncology (V.T. DeVita et al., eds., J.B. Lippincott Company, 2011).
II. Definitions id="p-39" id="p-39" id="p-39" id="p-39" id="p-39"
[0039] A "vector," as used herein, refers to a recombinant plasmid or virus that comprises a nucleic acid to be delivered into a host cell, either in vitro or in vivo. 259964/3 id="p-40" id="p-40" id="p-40" id="p-40" id="p-40"
[0040] The term "polynucleotide" or "nucleic acid" as used herein refers to a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides. Thus, this term includes, but is not limited to, single-, double- or multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, or a polymer comprising purine and pyrimidine bases, or other natural, chemically or biochemically modified, non-natural, or derivatized nucleotide bases. The backbone of the nucleic acid can comprise sugars and phosphate groups (as may typically be found in RNA or DNA), or modified or substituted sugar or phosphate groups.Alternatively, the backbone of the nucleic acid can comprise a polymer of synthetic subunits such as phosphoramidates and thus can be an oligodeoxynucleoside phosphoramidate (P-NH2) or a mixed phosphoramidate- phosphodiester oligomer. In addition, a double-stranded nucleic acid can be obtained from the single stranded polynucleotide product of chemical synthesis either by synthesizing the complementary strand and annealing the strands under appropriate conditions, or by synthesizing the complementary strand de novo using a DNA polymerase with an appropriate primer. id="p-41" id="p-41" id="p-41" id="p-41" id="p-41"
[0041] The terms "polypeptide" and "protein" are used interchangeably to refer to a polymer of amino acid residues, and are not limited to a minimum length. Such polymers of amino acid residues may contain natural or non-natural amino acid residues, and include, but are not limited to, peptides, oligopeptides, dimers, trimers, and multimers of amino acid residues. Both full-length proteins and fragments thereof are encompassed by the definition. The terms also include post-expression modifications of the polypeptide, for example, glycosylation, sialylation, acetylation, phosphorylation, and the like. Furthermore, for purposes of the present invention, a "polypeptide" refers to a protein which includes modifications, such as deletions, additions, and substitutions (generally conservative in nature), to the native sequence, as long as the protein maintains the desired activity. These modifications may be deliberate, as through site-directed mutagenesis, or may be accidental, such as through mutations of hosts which produce the proteins or errors due to PCR amplification. id="p-42" id="p-42" id="p-42" id="p-42" id="p-42"
[0042] A "recombinant viral vector" refers to a recombinant polynucleotide vector comprising one or more heterologous sequences (i.e., nucleic acid sequence not of viral origin). In the case of recombinant AAV vectors, the recombinant nucleic acid is flanked by at least one, e.g., two, inverted terminal repeat sequences (ITRs). 259964/3 id="p-43" id="p-43" id="p-43" id="p-43" id="p-43"
[0043] A "recombinant AAV vector (rAAV vector)" refers to a polynucleotide vector comprising one or more heterologous sequences (i.e., nucleic acid sequence not of AAV origin) that are flanked by at least one, e.g., two, AAV inverted terminal repeat sequences (ITRs). Such rAAV vectors can be replicated and packaged into infectious viral particles when present in a host cell that has been infected with a suitable helper virus (or that is expressing suitable helper functions) and that is expressing AAV rep and cap gene products (i.e. AAV Rep and Cap proteins). When a rAAV vector is incorporated into a larger polynucleotide (e.g., in a chromosome or in another vector such as a plasmid used for cloning or transfection), then the rAAV vector may be referred to as a "pro-vector" which can be "rescued" by replication and encapsidation in the presence of AAV packaging functions and suitable helper functions. A rAAV vector can be in any of a number of forms, including, but not limited to, plasmids, linear artificial chromosomes, complexed with lipids, encapsulated within liposomes, and, in embodiments, encapsidated in a viral particle, particularly an AAV particle. A rAAV vector can be packaged into an AAV virus capsid to generate a "recombinant adeno-associated viral particle (rAAV particle)". id="p-44" id="p-44" id="p-44" id="p-44" id="p-44"
[0044] An "rAAV virus" or "rAAV viral particle" refers to a viral particle composed of at least one AAV capsid protein and an encapsidated rAAV vector genome. id="p-45" id="p-45" id="p-45" id="p-45" id="p-45"
[0045] "Heterologous" means derived from a genotypically distinct entity from that of the rest of the entity to which it is compared or into which it is introduced or incorporated. For example, a nucleic acid introduced by genetic engineering techniques into a different cell type is a heterologous nucleic acid (and, when expressed, can encode a heterologous polypeptide). Similarly, a cellular sequence (e.g., a gene or portion thereof) that is incorporated into a viral vector is a heterologous nucleotide sequence with respect to the vector. id="p-46" id="p-46" id="p-46" id="p-46" id="p-46"
[0046] The term "transgene" refers to a nucleic acid that is introduced into a cell and is capable of being transcribed into RNA and optionally, translated and/or expressed under appropriate conditions. In aspects, it confers a desired property to a cell into which it was introduced, or otherwise leads to a desired therapeutic or diagnostic outcome. In another aspect, it may be transcribed into a molecule that mediates RNA interference, such as siRNA. 259964/3 id="p-47" id="p-47" id="p-47" id="p-47" id="p-47"
[0047] The terms "genome particles (gp)," "genome equivalents," or "genome copies" as used in reference to a viral titer, refer to the number of virions containing the recombinant AAV DNA genome, regardless of infectivity or functionality. The number of genome particles in a particular vector preparation can be measured by procedures such as described in the Examples herein, or for example, in Clark et al. (1999) Hum. Gene Ther., 10:1031-1039; Veldwrjk et al. (2002) Mol. Ther, 6:272-278. id="p-48" id="p-48" id="p-48" id="p-48" id="p-48"
[0048] The terms "infection unit (iu)," "infectious particle," or "replication unit," as used in reference to a viral titer, refer to the number of infectious and replication-competent recombinant AAV vector particles as measured by the infectious center assay, also known as replication center assay, as described, for example, in McLaughlin et al. (1988) J. Virol., 62:1963-1973. id="p-49" id="p-49" id="p-49" id="p-49" id="p-49"
[0049] The term "transducing unit (tu)" as used in reference to a viral titer, refers to the number of infectious recombinant AAV vector particles that result in the production of a functional transgene product as measured in functional assays such as described in Examples herein, or for example, in Xiao et al. (1997) Exp. Neurobiol., 144:113-124; or in Fisher et al. (1996) J. Virol, 70:520-532 (LFU assay). id="p-50" id="p-50" id="p-50" id="p-50" id="p-50"
[0050] An "inverted terminal repeat" or "ITR" sequence is a term well understood in the art and refers to relatively short sequences found at the termini of viral genomes which are in opposite orientation. id="p-51" id="p-51" id="p-51" id="p-51" id="p-51"
[0051] An "AAV inverted terminal repeat (ITR)" sequence, a term well-understood in the art, is an approximately 145-nucleotide sequence that is present at both termini of the native single-stranded AAV genome. The outermost 125 nucleotides of the ITR can be present in either of two alternative orientations, leading to heterogeneity between different AAV genomes and between the two ends of a single AAV genome. The outermost 125 nucleotides also contains several shorter regions of self-complementarity (designated A, A', B, B', C, C' and D regions), allowing intrastrand base-pairing to occur within this portion of the ITR. id="p-52" id="p-52" id="p-52" id="p-52" id="p-52"
[0052] A "terminal resolution sequence" or "trs" is a sequence in the D region of the AAV ITR that is cleaved by AAV rep proteins during viral DNA replication. A mutant terminal 259964/3 resolution sequence is refractory to cleavage by AAV rep proteins. "AAV helper functions" refer to functions that allow AAV to be replicated and packaged by a host cell. AAV helper functions can be provided in any of a number of forms, including, but not limited to, helper virus or helper virus genes which aid in AAV replication and packaging. Other AAV helper functions are known in the art such as genotoxic agents. id="p-53" id="p-53" id="p-53" id="p-53" id="p-53"
[0053] ‘ ‘AAV helper functions" refer to functions that allow AAV to be replicated andpackaged by a host cell. AAV helper functions can be provided in any of a number of forms, including, but not limited to, helper virus or helper virus genes which aid in AAV replication and packaging. Other AAV helper functions are known in the art such as genotoxic agents. id="p-54" id="p-54" id="p-54" id="p-54" id="p-54"
[0054] A "helper virus" for AAV refers to a virus that allows AAV (which is a defective parvovirus) to be replicated and packaged by a host cell. A number of such helper viruses have been identified, including adenoviruses, herpesviruses, poxviruses such as vaccinia, and baculovirus. The adenoviruses encompass a number of different subgroups, although Adenovirus type 5 of subgroup C (Ad5) is most commonly used. Numerous adenoviruses of human, non-human mammalian and avian origin are known and are available from depositories such as the ATCC. Viruses of the herpes family, which are also available from depositories such as ATCC, include, for example, herpes simplex viruses (HSV), Epstein-Barr viruses (EBV), cytomegaloviruses (CMV) and pseudorabies viruses (PRV). Baculoviruses available from depositories include Autographa californica nuclear polyhedrosis virus. id="p-55" id="p-55" id="p-55" id="p-55" id="p-55"
[0055] ‘Percent (%) sequence identity" with respect to a reference polypeptide or nucleicacid sequence is defined as the percentage of amino acid residues or nucleotides in a candidate sequence that are identical with the amino acid residues or nucleotides in the reference polypeptide or nucleic acid sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid or nucleic acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software programs, for example, those described in Current Protocols in Molecular Biology (Ausubel et al, eds., 1987), Supp. 30, section 7.7.18, Table 7.7.1, and including BLAST, 259964/3 BLAST-2, ALIGN or Megalign (DNASTAR) software. A potential alignment program is ALIGN Plus (Scientific and Educational Software, Pennsylvania). Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For purposes herein, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows: 100 times the fraction X/Y, where X is the number of amino acid residues scored as identical matches by the sequence alignment program in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. For purposes herein, the % nucleic acid sequence identity of a given nucleic acid sequence C to, with, or against a given nucleic acid sequence D (which can alternatively be phrased as a given nucleic acid sequence C that has or comprises a certain % nucleic acid sequence identity to, with, or against a given nucleic acid sequence D) is calculated as follows: 100 times the fraction W/Z, where W is the number of nucleotides scored as identical matches by the sequence alignment program in that program's alignment of C and D, and where Z is the total number of nucleotides in D. It will be appreciated that where the length of nucleic acid sequence C is not equal to the length of nucleic acid sequence D, the % nucleic acid sequence identity of C to D will not equal the % nucleic acid sequence identity of D to C. id="p-56" id="p-56" id="p-56" id="p-56" id="p-56"
[0056] An "isolated" molecule (e.g., nucleic acid or protein) or cell means it has been identified and separated and/or recovered from a component of its natural environment. id="p-57" id="p-57" id="p-57" id="p-57" id="p-57"
[0057] An "effective amount" is an amount sufficient to effect beneficial or desired results, including clinical results (e.g., amelioration of symptoms, achievement of clinical endpoints, and the like). An effective amount can be administered in one or more administrations. In terms of a disease state, an effective amount is an amount sufficient to ameliorate, stabilize, or delay development of a disease. For example, an effective amount of a rAAV particle 259964/3 expresses a desired amount of heterologous nucleic acid such as a therapeutic polypeptide or therapeutic nucleic acid. id="p-58" id="p-58" id="p-58" id="p-58" id="p-58"
[0058] An "individual" or "subject" is a mammal. Mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats). In certain embodiments, the individual or subject is a human. id="p-59" id="p-59" id="p-59" id="p-59" id="p-59"
[0059] As used herein, "treatment" is an approach for obtaining beneficial or desired clinical results. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (e.g., not worsening) state of disease, preventing spread (e.g., metastasis) of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment. id="p-60" id="p-60" id="p-60" id="p-60" id="p-60"
[0060] When used in reference to a gene or coding sequence, "N-acetylglucosamine-1- phosphotransferase (aka GlcNAc-1-phosphotransferase or GNPTAB)" refers to a polynucleotide sequence encoding the alpha and beta subunits of the enzyme that catalyzes a chemical reaction involving the formation of lysosomal-enzyme N-acetyl-glucosaminyl- phospho-D-mannose and UMP from UDP-N-acetyl-D-glucosamine and lysosomal-enzyme D- mannose (EC code 2.7.8.17). When used in reference to a polypeptide, "N-acetylglucosamine- 1-phosphotransferase (aka GlcNAc-1-phosphotransferase or GNPTAB)" refers to the alpha and beta subunits of aforementioned enzyme (Kudo, M. et al., JBiol Chem. 2005, 280(43):36141- 9; Gelfman, CM et al., Invest. Opthamol. Vis. Sci. 2007, 48(11):5221-5228). The full GlcNAc- 1-phosphotransferase enzyme complex is known to include a2, P2, and y2 subunits, of which the alpha and beta subunits are required for catalytic activity. Any enzyme known or predicted to catalyze the reaction described by EC code 2.7.8.17 and/or perform the molecular function described by GO term GO: 0003976 may be a GNPTAB of the present disclosure. In some embodiments, the GNPTAB is a variant GNPTAB. In some embodiments, the GNPTAB is a truncated GNPTAB. In some embodiments, nucleic acid encoding the GNPTAB is about 4.kb. In some embodiments, nucleic acid encoding the GNPTAB is less than about 4.7 kb. In 259964/3 some embodiments, the variant (e.g., truncated) GNPTAB comprises the alpha and beta subunits. In some embodiments, the variant GNPTAB (e.g., a truncated GNPTAB) is at least about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%, identical to native GNPTAB. In some embodiments, the variant GNPTAB (e.g., a truncated GNPTAB) maintains at least about 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, or 10% the activity of native GNPTAB. Examples of human GNPTAB are provided by GenBank Accession Nos. NP_077288.2 and NM_024312.4. An example of a human GNPTAB amino acid sequence is provided by SEQ ID NO:1. Examples of mouse GNPTAB are provided by GenBank Accession No. NP_001004164. An example of a mouse GNPTAB amino acid sequence is provided by SEQ ID NO:2. Additional examples of GNPTAB are provided by GenBank Accession Nos. XP_001155334 and XP_509312 (Pan troglodytes), XP_0026876and NP_001179157 (Bos taurus), XP_416329 (Gallus gallus), XP_532667 (Canis familiaris), XP_001497199 (Equus caballus), XP_001079967 and XP_343195 (Rattus norviegicus) and NP_001038233 (Danio rerio). id="p-61" id="p-61" id="p-61" id="p-61" id="p-61"
[0061] "Mucolipidosis type II" (the terms MLII, ML-II, and ML type II may be used interchangeably herein) and "mucolipidosis type III" (the terms MLIII, ML-III, and ML type III may be used interchangeably herein) refer to a class of diseases caused by mutations in the GNPTAB gene. Both diseases are autosomal recessive disorders. However, MLIII is typically associated with mutations causing a more mild loss of GNPTAB function as compared to MLII. As such, MLII typically results in more severe disease phenotypes than MLIII. MLII is also known as I-cell disease. Further description of MLII may be found in OMIM entry #252500. MLIII is also known as pseudo-Hurler polydystrophy. Further description of MLIII may be found in OMIM entry #252600. id="p-62" id="p-62" id="p-62" id="p-62" id="p-62"
[0062] " Chicken P-actin (CBA) promoter" refers to a polynucleotide sequence derived froma chicken P-actin gene (e.g., Gallus gallus beta actin, represented by GenBank Entrez Gene ID 396526). As used herein, "chicken P-actin promoter" may refer to a promoter containing a cytomegalovirus (CMV) early enhancer element, the promoter and first exon and intron of the chicken P-actin gene, and the splice acceptor of the rabbit beta-globin gene, such as the sequences described in Miyazaki, J., et al. (1989) Gene 79(2):269-77. As used herein, the term 259964/3 "CAG promoter" may be used interchangeably. As used herein, the term "CMV early enhancer/chicken beta actin (CAG) promoter" may be used interchangeably. id="p-63" id="p-63" id="p-63" id="p-63" id="p-63"
[0063] A shortened chicken beta actin promoter was chosen based on deletion studies that showed that sequences upstream of -106 could be deleted without significantly affecting promoter activity (Quitschke et al., J. Biol. Chem. 264:9539-9546, 1989). id="p-64" id="p-64" id="p-64" id="p-64" id="p-64"
[0064] Reference to "about" a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to "about X" includes description of "X." id="p-65" id="p-65" id="p-65" id="p-65" id="p-65"
[0065] As used herein, the singular form of the articles "a," "an," and "the" includes plural references unless indicated otherwise. id="p-66" id="p-66" id="p-66" id="p-66" id="p-66"
[0066] It is understood that aspects and embodiments of the invention described herein include "comprising," "consisting," and/or "consisting essentially of" aspects and embodiments.
III. Vectors id="p-67" id="p-67" id="p-67" id="p-67" id="p-67"
[0067] In certain aspects, the invention provides rAAV vectors, e.g., suitable for use in any of the methods, rAAV particles, and/or pharmaceutical compositions described herein. For example, in some embodiments, a heterologous nucleic acid (e.g., a polynucleotide sequence encoding a functional GNPTAB polypeptide) is delivered to a subject through a rAAV vector of the present disclosure. id="p-68" id="p-68" id="p-68" id="p-68" id="p-68"
[0068] Certain aspects of the present disclosure relate to N-acetylglucosamine-1-phosphate transferase, alpha and beta subunits (GNPTAB), e.g., GNPTAB polypeptides, or nucleic acids encoding a GNPTAB polypeptide. As is known in the art, the N-acetylglucosamine-1- phosphate transferase (also known as N-acetylglucosamine-1-phosphotransferase) enzyme includes two alpha, two beta, and two gamma subunits. The alpha and beta subunits are encoded by a GNPTAB gene (also known as GNPTA, I-cell disease or ICD, or EG432486 or mKIAA1208 in mouse). Examples of GNPTAB genes include, for example, human GNPTAB 259964/3 (e.g., as described by NCBI Gene ID No. 79158) and mouse GNPTAB (e.g., as described by NCBI Gene ID No. 432486). id="p-69" id="p-69" id="p-69" id="p-69" id="p-69"
[0069] In some embodiments, the GNPTAB polypeptide may be a human GNPTAB polypeptide. A human GNPTAB polypeptide sequence may include without limitation NCBI Reference Sequence No. NP_077288. In some embodiments, the GNPTAB polypeptide comprises the amino acid sequence of SEQ ID NO:1. In some embodiments, the GNPTAB polypeptide comprises an amino acid sequence that is at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to the amino acid sequence of SEQ ID NO:1. In some embodiments, the GNPTAB is a truncated GNPTAB. In some embodiments, nucleic acid encoding the GNPTAB is about 4.7 kb. In some embodiments, nucleic acid encoding the GNPTAB is less than about 4.7 kb. In some embodiments, the variant (e.g., truncated) GNPTAB comprises the alpha and beta subunits. In some embodiments, the GNPTAB polypeptide is a variant GNPTAB polypeptide (e.g., truncated GNPTAB) that maintains at least a portion of the activity of a wild-type GNPTAB polypeptide (e.g., at least about any of 5%, 10%, 25%, 50%, 75% or 100% of wild-type GNPTAB activity). In some embodiments, the variant GNPTAB polypeptide (e.g., truncated GNPTAB) has greater activity compared to a wild-type GNPTAB polypeptide (e.g., at least about any of 125%, 150%, 200%, 300%, or 500% greater activity compared to wild-type GNPTAB). id="p-70" id="p-70" id="p-70" id="p-70" id="p-70"
[0070] In some embodiments, the heterologous nucleic acid (e.g., nucleic acid encoding GNPTAB) is operably linked to a promoter. Exemplary promoters include, but are not limited to, the cytomegalovirus (CMV) immediate early promoter, the RSV LTR, the MoMLV LTR, the phosphoglycerate kinase- 1 (PGK) promoter, a simian virus 40 (SV40) promoter and a CKpromoter, a transthyretin promoter (TTR), a TK promoter, a tetracycline responsive promoter (TRE), an HBV promoter, an hAAT promoter, a LSP promoter, chimeric liver-specific promoters (LSPs), the E2F promoter, the telomerase (hTERT) promoter; the cytomegalovirus enhancer/chicken beta-actin/Rabbit P-globin promoter (CAG promoter; Niwa et al., Gene, 259964/3 1991, 108(2):193-9) and the elongation factor 1-alpha promoter (EFl-alpha) promoter (Kim et al., Gene, 1990, 91(2):217-23 and Guo etal., Gene Ther., 1996, 3(9):802-10). The promoter can be a constitutive, inducible or repressible promoter. In some embodiments, the promoter comprises a human P-glucuronidase promoter or a cytomegalovirus (CMV) enhancer linked to a chicken P-actin (CBA) promoter. In some embodiments, the promoter comprises a modified or truncated CBA promoter. In some embodiments, the promoter comprises a shortened CMV enhancer. id="p-71" id="p-71" id="p-71" id="p-71" id="p-71"
[0071] In some embodiments, the vector comprises an intron. For example, in some embodiments, the intron is a chimeric intron derived from chicken beta-actin and rabbit beta- globin. In some embodiments, the intron is a minute virus of mice (MVM) intron. id="p-72" id="p-72" id="p-72" id="p-72" id="p-72"
[0072] In some embodiments, the vector comprises a polyadenylation (polyA) sequence. Numerous examples of polyadenylation sequences are known in the art, such as a bovine growth hormone (BGH) Poly(A) sequence (see, e.g., accession number EF592533), an SVpolyadenylation sequence, and an HSV TK pA polyadenylation sequence. id="p-73" id="p-73" id="p-73" id="p-73" id="p-73"
[0073] Without wishing to be bound to theory, because of the large size of the GNPTAB coding sequence, it may be advantageous to minimize the size of other elements of the rAAV vector (e.g., a promoter, enhancer, intron, polyA sequence, and so forth). In some embodiments, a shortened variant of any of the promoters described herein may be used in a rAAV vector. Methods for generating a shortened variant of a promoter (e.g., a promoter listed above) are known in the art. For example, the promoter of interest could be mutated by introducing deletions and/or substitutions of one or more nucleotides in the promoter sequence, and such variant promoter sequences could be individually cloned into vectors that include a reporter construct under the regulation of each promoter sequence. This system could be used to identify shortened variants of a promoter that retain a designated strength (e.g., amount of transcript produced). In some embodiments, a rAAV vector of the present disclosure may include a modified, truncated, and/or shortened CMV enhancer/CBA promoter, e.g., as described herein. Similar methods could be used to identify shortened variants of an intron that retain proper levels of transcription, mRNA stability, and/or splicing. In some embodiments, a rAAV vector of the present disclosure may include a shortened intron as described herein. 259964/3 Similar methods could be used to identify shortened variants of a polyA sequence that retain proper levels of transcription, mRNA stability, and/or polyadenylation. In some embodiments, a rAAV vector of the present disclosure may include a shortened polyA sequence as described herein. In some embodiments the GTNAP gene comprises a nucleotide sequence encoding the amino acid sequence of SEQ ID NO:1 or 2. id="p-74" id="p-74" id="p-74" id="p-74" id="p-74"
[0074] The present invention contemplates the use of a recombinant viral genome for introduction of one or more nucleic acid sequences encoding a therapeutic polypeptide and/or nucleic acid for packaging into a rAAV viral particle. The recombinant viral genome may include any element to establish the expression of the therapeutic polypeptide and/or nucleic acid, for example, a promoter, an ITR of the present disclosure, a ribosome binding element, terminator, enhancer, selection marker, intron, polyA signal, and/or origin of replication.
IV. Viral particles and methods of producing viral particles id="p-75" id="p-75" id="p-75" id="p-75" id="p-75"
[0075] Certain aspects of the present disclosure relate to rAAV particles, e.g., containing a rAAV vector of the present disclosure. In an AAV particle, a nucleic acid is encapsidated in the AAV particle. The AAV particle also comprises capsid proteins. In some embodiments, the nucleic acid comprises the coding sequence(s) of interest (e.g., a GNPTAB coding sequence) operatively linked components in the direction of transcription, control sequences including transcription initiation and termination sequences, thereby forming an expression cassette. The expression cassette is flanked on the 5' and 3' end by at least one functional AAV ITR sequence. By "functional AAV ITR sequences" it is meant that the ITR sequences function as intended for the rescue, replication and packaging of the AAV virion. See Davidson et al, PNAS, 2000, 97(7)3428-32; Passini et al, J. Virol., 2003, 77(12):7034-40; and Pechan et al., Gene Ther., 2009, 16:10-16. For practicing some aspects of the invention, the recombinant vectors comprise at least all of the sequences of AAV essential for encapsidation and the physical structures for infection by the rAAV. AAV ITRs for use in the vectors of the invention need not have a wild-type nucleotide sequence (e.g., as described in Kotin, Hum. Gene Ther., 1994, 5:793-801), and may be altered by the insertion, deletion or substitution of nucleotides or the AAV ITRs may be derived from any of several AAV serotypes. More than serotypes of AAV are currently 259964/3 known, and new serotypes and variants of existing serotypes continue to be identified. See Gao et al, PNAS, 2002, 99(18): 11854-6; Gao et al, PNAS, 2003, 100(10):6081-6; and Bossis et al., J. Virol., 2003, 77(12):6799-810. Use of any AAV serotype is considered within the scope of the present invention. In some embodiments, a rAAV vector is a vector derived from an AAV serotype, including without limitation, AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAVrh8R, AAV9, AAV10, AAVrh10, AAV11, AAV12, AAV2R471A, AAV DJ, a goat AAV, bovine AAV, or mouse AAV ITRs or the like. In some embodiments, the nucleic acid in the AAV comprises an ITR of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAVrh8R, AAV9, AAV10, AAVrh10, AAV11, AAV12, AAV2R471A, AAV DJ, a goat AAV, bovine AAV, or mouse AAV ITRs or the like. id="p-76" id="p-76" id="p-76" id="p-76" id="p-76"
[0076] In some embodiments, a rAAV particle comprises an encapsidation protein selected from AAV1, AAV2, AAV3, AAV4, AAV5, AAV6 (e.g., a wild-type AAV6 capsid, or a variant AAV6 capsid such as ShH10, as described in U.S. PG Pub. 2012/0164106), AAV7, AAV8, AAVrh8, AAVrh8R, AAV9 (e.g., a wild-type AAV9 capsid, or a modified AAVcapsid as described in U.S. PG Pub. 2013/0323226), AAV10, AAVrh10, AAV11, AAV12, a tyrosine capsid mutant, a heparin binding capsid mutant, an AAV2R471A capsid, an AAVAAV2/2-7m8 capsid, an AAV DJ capsid (e.g., an AAV-DJ/8 capsid, an AAV-DJ/capsid, or any other of the capsids described in U.S. PG Pub. 2012/0066783), AAV2 N587A capsid, AAV2 E548A capsid, AAV2 N708A capsid, AAV V708K capsid, goat AAV capsid, AAV1/AAV2 chimeric capsid, bovine AAV capsid, mouse AAV capsid, rAAV2/HBoVcapsid, or an AAV capsid described in U.S. Pat. No. 8,283,151 or International Publication No. WO/2003/042397. In further embodiments, a rAAV particle comprises capsid proteins of an AAV serotype from Clades A-F. id="p-77" id="p-77" id="p-77" id="p-77" id="p-77"
[0077] Different AAV serotypes are used to optimize transduction of particular target cells or to target specific cell types within a particular target tissue (e.g., a diseased tissue). A rAAV particle can comprise viral proteins and viral nucleic acids of the same serotype or a mixed serotype. For example, a rAAV particle may contain one or more ITRs and capsid derived from the same AAV serotype, or a rAAV particle may contain one or more ITRs derived from a different AAV serotype than capsid of the rAAV particle. In certain embodiments, a rAAV particle contains an AAV8 capsid and one or more (e.g., 2) AAV2 ITRs. 259964/3 Production of AAV particles id="p-78" id="p-78" id="p-78" id="p-78" id="p-78"
[0078] Numerous methods are known in the art for production of rAAV vectors, including transfection, stable cell line production, and infectious hybrid virus production systems which include adenovirus-AAV hybrids, herpesvirus-AAV hybrids (Conway, JE et al., (1997) J. Virology 71(11):8780-8789) and baculovirus-AAV hybrids (Urabe, M. et al., (2002) Human Gene Therapy 13(16):1935-1943; Kotin, R. (2011) Hum Mol Genet. 20(R1): R2-R6). rAAV production cultures for the production of rAAV virus particles all require; 1) suitable host cells, 2) suitable helper virus function, 3) AAV rep and cap genes and gene products; 4) a nucleic acid (such as a therapeutic nucleic acid) flanked by at least one AAV ITR sequences (e.g., an AAV genome encoding GNPTAB); and 5) suitable media and media components to support rAAV production. In some embodiments, the suitable host cell is a primate host cell. In some embodiments, the suitable host cell is a human-derived cell lines such as HeLa, A549, 293, or Perc.6 cells. In some embodiments, the suitable helper virus function is provided by wild-type or mutant adenovirus (such as temperature sensitive adenovirus), herpes virus (HSV), baculovirus, or a plasmid construct providing helper functions. In some embodiments, the AAV rep and cap gene products may be from any AAV serotype. In general, but not obligatory, the AAV rep gene product is of the same serotype as the ITRs of the rAAV vector genome as long as the rep gene products may function to replicated and package the rAAV genome. Suitable media known in the art may be used for the production of rAAV vectors. These media include, without limitation, media produced by Hyclone Laboratories and JRH including Modified Eagle Medium (MEM), Dulbecco's Modified Eagle Medium (DMEM), custom formulations such as those described in U.S. Patent No. 6,566,118, and Sf-900 II SFM media as described in U.S. Patent No. 6,723,551, particularly with respect to custom media formulations for use in production of recombinant AAV vectors. In some embodiments, the AAV helper functions are provided by adenovirus or HSV. In some embodiments, the AAV helper functions are provided by baculovirus and the host cell is an insect cell (e.g., Spodoptera frugiperda (Sf9) cells). id="p-79" id="p-79" id="p-79" id="p-79" id="p-79"
[0079] One method for producing rAAV particles is the triple transfection method. Briefly, a plasmid containing a rep gene and a capsid gene, along with a helper adenoviral plasmid, may 259964/3 be transfected (e.g., using the calcium phosphate method) into a cell line (e.g., HEK-2cells), and virus may be collected and optionally purified. As such, in some embodiments, the rAAV particle was produced by triple transfection of a nucleic acid encoding the rAAV vector, a nucleic acid encoding AAV rep and cap, and a nucleic acid encoding AAV helper virus functions into a host cell, wherein the transfection of the nucleic acids to the host cells generates a host cell capable of producing rAAV particles. id="p-80" id="p-80" id="p-80" id="p-80" id="p-80"
[0080] In some embodiments, rAAV particles may be produced by a producer cell line method (see Martin et al., (2013) Human Gene Therapy Methods 24:253-269; U.S. PG Pub. No. US2004/0224411; and Liu, X.L. et al. (1999) Gene Ther. 6:293-299). Briefly, a cell line (e.g., a HeLa, 293, A549, or Perc.6 cell line) may be stably transfected with a plasmid containing a rep gene, a capsid gene, and a vector genome comprising a promoter-heterologous nucleic acid sequence (e.g., GNPTAB). Cell lines may be screened to select a lead clone for rAAV production, which may then be expanded to a production bioreactor and infected with a helper virus (e.g., an adenovirus or HSV) to initiate rAAV production. Virus may subsequently be harvested, adenovirus may be inactivated (e.g., by heat) and/or removed, and the rAAV particles may be purified. As such, in some embodiments, the rAAV particle was produced by a producer cell line comprising one or more of nucleic acid encoding the rAAV vector, a nucleic acid encoding AAV rep and cap, and a nucleic acid encoding AAV helper virus functions. As described herein, the producer cell line method may be advantageous for the production of rAAV particles with an oversized genome, as compared to the triple transfection method. id="p-81" id="p-81" id="p-81" id="p-81" id="p-81"
[0081] In some embodiments, the nucleic acid encoding AAV rep and cap genes and/or the rAAV genome are stably maintained in the producer cell line. In some embodiments, nucleic acid encoding AAV rep and cap genes and/or the rAAV genome is introduced on one or more plasmids into a cell line to generate a producer cell line. In some embodiments, the AAV rep, AAV cap, and rAAV genome are introduced into a cell on the same plasmid. In other embodiments, the AAV rep, AAV cap, and rAAV genome are introduced into a cell on different plasmids. In some embodiments, a cell line stably transfected with a plasmid maintains the plasmid for multiple passages of the cell line (e.g., 5, 10, 20, 30, 40, 50 or more than 50 passages of the cell). For example, the plasmid(s) may replicate as the cell replicates, 259964/3 or the plasmid(s) may integrate into the cell genome. A variety of sequences that enable a plasmid to replicate autonomously in a cell (e.g., a human cell) have been identified (see, e.g., Krysan, P.J. et al. (1989) Mol. Cell Biol. 9:1026-1033). In some embodiments, the plasmid(s) may contain a selectable marker (e.g., an antibiotic resistance marker) that allows for selection of cells maintaining the plasmid. Selectable markers commonly used in mammalian cells include without limitation blasticidin, G418, hygromycin B, zeocin, puromycin, and derivatives thereof. Methods for introducing nucleic acids into a cell are known in the art and include without limitation viral transduction, cationic transfection (e.g., using a cationic polymer such as DEAE-dextran or a cationic lipid such as lipofectamine), calcium phosphate transfection, microinjection, particle bombardment, electroporation, and nanoparticle transfection (for more details, see e.g., Kim, T.K. and Eberwine, J.H. (2010) Anal. Bioanal. Chem. 397:3173-3178). id="p-82" id="p-82" id="p-82" id="p-82" id="p-82"
[0082] In some embodiments, the nucleic acid encoding AAV rep and cap genes and/or the rAAV genome are stably integrated into the genome of the producer cell line. In some embodiments, nucleic acid encoding AAV rep and cap genes and/or the rAAV genome is introduced on one or more plasmids into a cell line to generate a producer cell line. In some embodiments, the AAV rep, AAV cap, and rAAV genome are introduced into a cell on the same plasmid. In other embodiments, the AAV rep, AAV cap, and rAAV genome are introduced into a cell on different plasmids. In some embodiments, the plasmid(s) may contain a selectable marker (e.g., an antibiotic resistance marker) that allows for selection of cells maintaining the plasmid. Methods for stable integration of nucleic acids into a variety of host cell lines are known in the art. For example, repeated selection (e.g., through use of a selectable marker) may be used to select for cells that have integrated a nucleic acid containing a selectable marker (and AAV cap and rep genes and/or a rAAV genome). In other embodiments, nucleic acids may be integrated in a site-specific manner into a cell line to generate a producer cell line. Several site-specific recombination systems are known in the art, such as FLP/FRT (see, e.g., O'Gorman, S. et al. (1991) Science 251:1351-1355), Cre/loxP (see, e.g., Sauer, B. and Henderson, N. (1988) Proc. Natl. Acad. Sci. 85:5166-5170), and phi C31-att (see, e.g., Groth, A.C. et al. (2000) Proc. Natl. Acad. Sci. 97:5995-6000). 259964/3 id="p-83" id="p-83" id="p-83" id="p-83" id="p-83"
[0083] In some embodiments, the producer cell line is derived from a primate cell line (e.g., a non-human primate cell line, such as a Vero or FRhL-2 cell line). In some embodiments, the cell line is derived from a human cell line. In some embodiments, the producer cell line is derived from HeLa, 293, A549, or PERC.6® (Crucell) cells. For example, prior to introduction and/or stable maintenance/integration of nucleic acid encoding AAV rep and cap genes and/or the oversized rAAV genome into a cell line to generate a producer cell line, the cell line is a HeLa, 293, A549, or PERC.6® (Crucell) cell line, or a derivative thereof. id="p-84" id="p-84" id="p-84" id="p-84" id="p-84"
[0084] In some embodiments, the producer cell line is adapted for growth in suspension. As is known in the art, anchorage-dependent cells are typically not able to grow in suspension without a substrate, such as microcarrier beads. Adapting a cell line to grow in suspension may include, for example, growing the cell line in a spinner culture with a stirring paddle, using a culture medium that lacks calcium and magnesium ions to prevent clumping (and optionally an antifoaming agent), using a culture vessel coated with a siliconizing compound, and selecting cells in the culture (rather than in large clumps or on the sides of the vessel) at each passage. For further description, see, e.g., ATCC frequently asked questions document (available at www.atcc.org/Global/FAQs/9/1/Adapting%20a%20monolayer%20cell%20line%20to%20susp ension-40.aspx) and references cited therein. id="p-85" id="p-85" id="p-85" id="p-85" id="p-85"
[0085] In some aspects, a method is provided for producing any rAAV particle as disclosed herein comprising (a) culturing a host cell under a condition that rAAV particles are produced, wherein the host cell comprises (i) one or more AAV package genes, wherein each said AAV packaging gene encodes an AAV replication and/or encapsidation protein; (ii) a rAAV pro­vector comprising a nucleic acid encoding a heterologous nucleic acid as described herein flanked by at least one AAV ITR, and (iii) an AAV helper function; and (b) recovering the rAAV particles produced by the host cell. In some embodiments, said at least one AAV ITR is selected from the group consisting of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAVrh8R, AAV9, AAV10, AAVrh10, AAV11, AAV12, AAV2R471A, AAV DJ, a goat AAV, bovine AAV, or mouse AAV serotype ITRs or the like. For example, in some embodiments, the AAV serotype is AAV1, AAV2, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAVrh8R, AAV9, AAV10, or AAVrh10. In certain embodiments, the nucleic acid in the AAV comprises an AAV2 ITR. In some embodiments, said encapsidation protein is 259964/3 selected from the group consisting of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAVrh8R, AAV9, AAV10, AAVrh10, AAV11, AAV12, AAV2R471A, AAV2/2-7m8, AAV DJ, AAV2 N587A, AAV2 E548A, AAV2 N708A, AAV V708K, goat AAV, AAV1/AAV2 chimeric, bovine AAV, or mouse AAV capsid rAAV2/HBoV1 serotype capsid proteins or mutants thereof. In some embodiments, the encapsidation protein is an AAV8 capsid protein. In some embodiments, the rAAV particles comprise an AAV8 capsid and a recombinant genome comprising AAV2 ITRs, and nucleic acid encoding a therapeutic transgene/nucleic acid (e.g., nucleic acid encoding GNPTAB). id="p-86" id="p-86" id="p-86" id="p-86" id="p-86"
[0086] Suitable rAAV production culture media of the present invention may be supplemented with serum or serum-derived recombinant proteins at a level of 0.5%-20% (v/v or w/v). Alternatively, as is known in the art, rAAV vectors may be produced in serum-free conditions which may also be referred to as media with no animal-derived products. One of ordinary skill in the art may appreciate that commercial or custom media designed to support production of rAAV vectors may also be supplemented with one or more cell culture components know in the art, including without limitation glucose, vitamins, amino acids, and or growth factors, in order to increase the titer of rAAV in production cultures. id="p-87" id="p-87" id="p-87" id="p-87" id="p-87"
[0087] rAAV production cultures can be grown under a variety of conditions (over a wide temperature range, for varying lengths of time, and the like) suitable to the particular host cell being utilized. As is known in the art, rAAV production cultures include attachment- dependent cultures which can be cultured in suitable attachment-dependent vessels such as, for example, roller bottles, hollow fiber filters, microcarriers, and packed-bed or fluidized-bed bioreactors. rAAV vector production cultures may also include suspension-adapted host cells such as HeLa, 293, and SF-9 cells which can be cultured in a variety of ways including, for example, spinner flasks, stirred tank bioreactors, and disposable systems such as the Wave bag system. id="p-88" id="p-88" id="p-88" id="p-88" id="p-88"
[0088] rAAV vector particles of the invention may be harvested from rAAV production cultures by lysis of the host cells of the production culture or by harvest of the spent media from the production culture, provided the cells are cultured under conditions known in the art to cause release of rAAV particles into the media from intact cells, as described more fully in 259964/3 U.S. Patent No. 6,566,118). Suitable methods of lysing cells are also known in the art and include for example multiple freeze/thaw cycles, sonication, microfluidization, and treatment with chemicals, such as detergents and/or proteases. id="p-89" id="p-89" id="p-89" id="p-89" id="p-89"
[0089] In a further embodiment, the rAAV particles are purified. The term "purified" as used herein includes a preparation of rAAV particles devoid of at least some of the other components that may also be present where the rAAV particles naturally occur or are initially prepared from. Thus, for example, isolated rAAV particles may be prepared using a purification technique to enrich it from a source mixture, such as a culture lysate or production culture supernatant. Enrichment can be measured in a variety of ways, such as, for example, by the proportion of DNase-resistant particles (DRPs) or genome copies (gc) present in a solution, or by infectivity, or it can be measured in relation to a second, potentially interfering substance present in the source mixture, such as contaminants, including production culture contaminants or in-process contaminants, including helper virus, media components, and the like. id="p-90" id="p-90" id="p-90" id="p-90" id="p-90"
[0090] In some embodiments, the rAAV production culture harvest is clarified to remove host cell debris. In some embodiments, the production culture harvest is clarified by filtration through a series of depth filters including, for example, a grade DOHC Millipore Millistak+ HC Pod Filter, a grade A1HC Millipore Millistak+ HC Pod Filter, and a 0.2 p,m Filter Opticap XL1O Millipore Express SHC Hydrophilic Membrane filter. Clarification can also be achieved by a variety of other standard techniques known in the art, such as, centrifugation or filtration through any cellulose acetate filter of 0.2 p,m or greater pore size known in the art. id="p-91" id="p-91" id="p-91" id="p-91" id="p-91"
[0091] In some embodiments, the rAAV production culture harvest is further treated with Benzonase® to digest any high molecular weight DNA present in the production culture. In some embodiments, the Benzonase® digestion is performed under standard conditions known in the art including, for example, a final concentration of 1-2.5 units/ml of Benzonase® at a temperature ranging from ambient to 37°C for a period of 30 minutes to several hours. id="p-92" id="p-92" id="p-92" id="p-92" id="p-92"
[0092] rAAV particles may be isolated or purified using one or more of the following purification steps: equilibrium centrifugation; flow-through anionic exchange filtration; tangential flow filtration (TFF) for concentrating the rAAV particles; rAAV capture by apatite 259964/3 chromatography; heat inactivation of helper virus; rAAV capture by hydrophobic interaction chromatography; buffer exchange by size exclusion chromatography (SEC); nanofiltration; and rAAV capture by anionic exchange chromatography, cationic exchange chromatography, or affinity chromatography. These steps may be used alone, in various combinations, or in different orders. In some embodiments, the method comprises all the steps in the order as described below. Methods to purify rAAV particles are found, for example, in Xiao et al., (1998) Journal of Virology 72:2224-2232; US Patent Numbers 6,989,264 and 8,137,948; and WO 2010/148143.
V. Methods of Treatment id="p-93" id="p-93" id="p-93" id="p-93" id="p-93"
[0093] Certain aspects of the present disclosure relate to methods of treating mucolipidosis type II and/or mucolipidosis type III or increasing body size, increasing bone mineral content, or increasing bone mineral density in a mammal with mucolipidosis type II or mucolipidosis type III. These methods are based in part on the discovery described herein that AAV- mediated expression of GNPTAB can ameliorate symptoms of MLII, such as bone growth impairments, in a mouse model of the disease. As described above, both diseases are caused by loss-of-function mutations in the GNPTAB gene, which encodes the catalytic alpha and beta subunits of GlcNAc-1-phosphotransferase. id="p-94" id="p-94" id="p-94" id="p-94" id="p-94"
[0094] MLII is known to be an autosomal recessive disorder caused by mutations in GNPTAB. GNPTAB activity is required to add mannose-6-phosphate to proteins, thereby marking them for trafficking to the lysosome. In the absence of GNPTAB activity, lysosomal proteins (e.g., lysosomal hydrolases) are instead secreted extracellularly. As a result, substances that are normally broken down in lysosomes, such as glycosaminoglycans, lipids, and oligosaccharides, accumulate in cells, leading to the presence of large inclusions. MLII has also been called I-cell disease, due to the presence of these inclusion-cells ("I-cells"), which are identifiable by microscope. The symptoms of MLII often present shortly after birth and may include skeletal abnormalities, short stature, weak muscle tone, lack of muscle tone (hypotonia), hernias (e.g., protruded abdomen, umbilical hernias), hip dislocation and joint deformities, cardiomegaly, thickening and insufficiency of the mitral valve, progressive mucosal thickening of the airways, coarse and/or noisy breathing, frequent respiratory 259964/3 infections, constipation, diarrhea, delays in the development of gross and fine motor skills, hearing loss, and developmental delays, particularly in speech and motor skills, cognitive deficits. These symptoms are debilitating for MLII patients, who typically are not able to walk independently and do not survive past childhood. id="p-95" id="p-95" id="p-95" id="p-95" id="p-95"
[0095] Like MLII, MLIII is known in the art as an autosomal recessive disorder caused by mutations in GNPTAB. However, compared with MLII, MLIII typically results from weaker loss-of-function mutations in GNPTAB and is thereby characterized by milder disease phenotypes. MLIII symptoms may not be fully apparent until 3-5 years of age and are quite variable in severity, with some patients living past 60 while others do not survive past childhood. MLIII symptoms typically include skeletal abnormalities, short stature, aortic valve disease, corneal clouding, mild organ enlargement, loss of mobility, and joint abnormalities. MLIII has also been called pseudo-Hurler polydistrophy. For more detailed description and specific mutations found in MLII and MLIII patients, see, e.g., Paik, K.H., etal. (2005) Hum. Mutat. 26(4):308-14. id="p-96" id="p-96" id="p-96" id="p-96" id="p-96"
[0096] Various diagnostic tests for MLII and MLIII are known in the art. In some embodiments, MLII and/or MLIII may be diagnosed by measuring the activity of one or more lysosomal enzymes in a serum sample or other patient sample (e.g., a skin sample or cultured fibroblasts). The activity of certain lysosomal enzymes in serum may be 5- to 20-fold higher in MLII patients than in normal patients. Similarly, in MLIII the serum activity of these enzymes may be elevated up to 10-fold. These enzymes may include without limitation beta-D- hexosaminidase (EC code 3.2.1.52), beta-D-glucuronidase (EC code 3.2.1.31), beta-D- galactosidase (EC code 3.2.1.23), alpha-L-fucosidase (EC code 3.2.1.51) and alpha-D- mannosidase (EC code 3.2.1.24). In contrast, these activities in cultured fibroblasts may be deficient. In some embodiments, N-acetylglucosamine-1-phosphotransferase activity may be measured in order to diagnose MLII or MLIII. In some embodiments, a patient whose sample shows an N-acetylglucosamine-1-phosphotransferase activity of less than 1% compared to activity from a normal patient sample may be diagnosed with MLII. In some embodiments, a patient whose sample shows an N-acetylglucosamine-1-phosphotransferase activity of between 1%-10% compared to activity from a normal patient sample may be diagnosed with MLII. 259964/3 id="p-97" id="p-97" id="p-97" id="p-97" id="p-97"
[0097] In some embodiments, MLII and/or MLIII may be diagnosed by sequencing the GNPTAB locus (e.g., the coding sequence, promoter/intron sequences, or any other control sequences) for mutations. MLII and MLIII may also be characterized by increased urinary polysaccharides and/or urinary glycosaminoglycans, but these symptoms may be not specific to MLII and MLIII. In some embodiments, MLII and/or MLIII may be diagnosed prenatally by examining a chorionic sample, e.g., a trophoblast biopsy (see, e.g., Poenaru, L., et al. (1984)Am. J. Hum. Genet. 36(6):1379-85). As described above, in MLII, I-cells from a patient sample (e.g., fibroblasts) may also be identified by microscopy. id="p-98" id="p-98" id="p-98" id="p-98" id="p-98"
[0098] In some embodiments, a treatment for MLII and/or MLIII (e.g., a gene therapy vector of the present disclosure) may be tested for therapeutic efficacy. For example, in some embodiments, a treatment for MLII and/or MLIII may result in an increase in body height gain, bone mineral content, bone mineral density, or may result in an amelioration of one or more symptoms of MLII and/or MLIII as described herein. The effectiveness of rAAV administration can be monitored by several criteria as described herein. For example, after treatment in a subject using methods of the present invention, the subject may be assessed for e.g., an improvement and/or stabilization and/or delay in the progression of one or more signs or symptoms of the disease state by one or more clinical parameters including those described herein. Examples of such tests are known in the art, and include objective as well as subjective (e.g., subject reported) measures. id="p-99" id="p-99" id="p-99" id="p-99" id="p-99"
[0099] In some embodiments, expression of GNPTAB may be measured to monitor therapeutic efficacy. Measuring GNPTAB expression may refer to measuring GNPTAB mRNA and/or protein expression. Various methods for measuring mRNA and/or protein expression are known in the art, including without limitation qPCR, Northern blotting, RNA- seq, semi-quantitative PCR, Western blotting, mass spectrometry, ELISA, and so forth. id="p-100" id="p-100" id="p-100" id="p-100" id="p-100"
[0100] In some embodiments, the activity of one or more lysosomal enzymes, including without limitation beta-D-hexosaminidase (EC code 3.2.1.52), beta-D-glucuronidase (EC code 3.2.1.31), beta-D-galactosidase (EC code 3.2.1.23), alpha-L-fucosidase (EC code 3.2.1.51) and alpha-D- mannosidase (EC code 3.2.1.24), may be measured in a patient sample (e.g., a serum sample) to 259964/3 monitor therapeutic efficacy. A decrease in lysosomal enzyme activity in serum may indicate efficacy. id="p-101" id="p-101" id="p-101" id="p-101" id="p-101"
[0101] In some embodiments, an improvement in joint or limb function may indicate efficacy.In some embodiments, an improvement in speech may indicate efficacy. In some embodiments, an improvement in motor function may indicate efficacy. In some embodiments, increased growth rate (e.g., an increase in body height gain) may indicate efficacy. In some embodiments, as described in the Examples herein, bone mineral density, bone mineral content, and/or growth (e.g., height) may be assessed for monitoring therapeutic efficacy. Methods for monitoring height are well known to one of skill in the art. Tests for monitoring bone mineral density and bone mineral content (e.g., bone densitometry tests) are also well known to one of skill in the art (e.g., as described herein) and may include without limitation dual-energy x-ray absorptiometry (DEXA) scan, peripheral dual-energy x-ray absorptiometry (P-DEXA) scan, dual photon absorptiometry (DPA), and a computed tomography (CT) scan. id="p-102" id="p-102" id="p-102" id="p-102" id="p-102"
[0102] In some embodiments, a treatment for MLII and/or MLIII (e.g., a gene therapy vector of the present disclosure) may be tested in an animal model. Animal models for MLII and MLIII are known in the art. In some embodiments, a treatment for MLII and/or MLIII may be tested in a mouse model, such as the genetically modified mouse model described in the Examples herein. In some embodiments, a treatment for MLII may be tested in a feline model for MLII (see Mazrier, H., et al. (2003) J. Hered. 94(5):363-73 or Bosshard, N.U., et al. (1996) Vet. Pathol. 33(1):1-13 for further descriptions). id="p-103" id="p-103" id="p-103" id="p-103" id="p-103"
[0103] The selection of a particular rAAV vector and composition depend on a number of different factors, including, but not limited to, the individual human’s medical history and features of the condition and the individual being treated. The assessment of such features and the design of an appropriate therapeutic regimen is ultimately the responsibility of the prescribing physician. id="p-104" id="p-104" id="p-104" id="p-104" id="p-104"
[0104] In some aspects, the invention provides methods of treating MLII and/or MLIII by administering an effective amount of a rAAV particle of the present disclosure. rAAV may be administered to a particular tissue of interest, or it may be administered systemically. In some embodiments, an effective amount of rAAV may be administered parenterally. Parenteral routes 259964/3 of administration may include without limitation intravenous, intraperitoneal, intraosseous, intra­arterial, intracerebral, intramuscular, intrathecal, subcutaneous, intracerebroventricular, intrahepatic, and so forth. In some embodiments, an effective amount of rAAV may be administered through one route of administration. In some embodiments, an effective amount of rAAV may be administered through a combination of more than one route of administration. In some embodiments, an effective amount of rAAV is administered to one location. In other embodiments, an effective amount of rAAV may be administered to more than one location. id="p-105" id="p-105" id="p-105" id="p-105" id="p-105"
[0105] An effective amount of rAAV (in some embodiments in the form of particles) is administered, depending on the objectives of treatment. For example, where a low percentage of transduction can achieve the desired therapeutic effect, then the objective of treatment is generally to meet or exceed this level of transduction. In some instances, this level of transduction can be achieved by transduction of only about 1 to 5% of the target cells of the desired tissue type, in some embodiments at least about 20% of the cells of the desired tissue type, in some embodiments at least about 50%, in some embodiments at least about 80%, in some embodiments at least about 95%, in some embodiments at least about 99% of the cells of the desired tissue type. The rAAV composition may be administered by one or more administrations, either during the same procedure or spaced apart by days, weeks, months, or years. One or more of any of the routes of administration described herein may be used. In some embodiments, multiple vectors may be used to treat the human. id="p-106" id="p-106" id="p-106" id="p-106" id="p-106"
[0106] Methods to identify cells transduced by AAV viral particles are known in the art; for example, immunohistochemistry or the use of a marker such as enhanced green fluorescent protein can be used to detect transduction of viral particles; for example viral particles comprising a rAAV capsid with one or more substitutions of amino acids. id="p-107" id="p-107" id="p-107" id="p-107" id="p-107"
[0107] In some embodiments an effective amount of rAAV particles is administered to more than one location simultaneously or sequentially. In other embodiments, an effective amount of rAAV particles is administered to a single location more than once (e.g., repeated). In some embodiments, multiple injections of rAAV viral particles are no more than one hour, two hours, three hours, four hours, five hours, six hours, nine hours, twelve hours or 24 hours apart. 259964/3 id="p-108" id="p-108" id="p-108" id="p-108" id="p-108"
[0108] An effective amount of rAAV (in some embodiments in the form of particles) is administered, depending on the objectives of treatment. For example, where a low percentage of transduction can achieve the desired therapeutic effect, then the objective of treatment is generally to meet or exceed this level of transduction. In some instances, this level of transduction can be achieved by transduction of only about 1 to 5% of the target cells, in some embodiments at least about 20% of the cells of the desired tissue type, in some embodiments at least about 50%, in some embodiments at least about 80%, in some embodiments at least about 95%, in some embodiments at least about 99% of the cells of the desired tissue type. The rAAV composition may be administered by one or more administrations, either during the same procedure or spaced apart by days, weeks, months, or years. In some embodiments, multiple vectors may be used to treat the mammal (e.g., a human). id="p-109" id="p-109" id="p-109" id="p-109" id="p-109"
[0109] In some embodiments, a rAAV composition of the present disclosure may be used for administration to a human. In some embodiments, a rAAV composition of the present disclosure may be used for pediatric administration. Without wishing to be bound to theory, because many of the symptoms of MLII and MLIII are developmental in nature (e.g., growth, limb, and joint defects; speech and motor delays), it may be particularly advantageous to treat MLII and/or MLIII as early in life as possible. In some embodiments, an effective amount of rAAV (in some embodiments in the form of particles) is administered to a patient that is less than one month, less than two months, less than three months, less than four months, less than five months, less than six months, less than seven months, less than eight months, less than nine months, less than ten months, less than eleven months, less than one year, less than 13 months, less than months, less than 15 months, less than 16 months, less than 17 months, less than 18 months, less than 19 months, less than 20 months, less than 21 months, less than 22 months, less than two years, or less than three years old. id="p-110" id="p-110" id="p-110" id="p-110" id="p-110"
[0110] In some embodiments, a rAAV composition of the present disclosure may be used for administration to a young adult. In some embodiments, an effective amount of rAAV (in some embodiments in the form of particles) is administered to a patient that is less than 12 years old, less than 13 years old, less than 14 years old, less than 15 years old, less than 16 years old, less than 17 years old, less than 18 years old, less than 19 years old, less than 20 years old, less than 259964/3 21 years old, less than 22 years old, less than 23 years old, less than 24 years old, or less than years old.
VI. Kits or Articles of Manufacture id="p-111" id="p-111" id="p-111" id="p-111" id="p-111"
[0111] The rAAV vectors, particles, and/or pharmaceutical compositions as described herein may be contained within a kit or article of manufacture, e.g., designed for use in one of the methods of the invention as described herein. id="p-112" id="p-112" id="p-112" id="p-112" id="p-112"
[0112] Generally, the system comprises a cannula, one or more syringes (e.g., 1, 2, 3, 4 or more), and one or more fluids (e.g., 1, 2, 3, 4 or more) suitable for use in the methods of the invention. id="p-113" id="p-113" id="p-113" id="p-113" id="p-113"
[0113] The syringe may be any suitable syringe, provided it is capable of being connected to the cannula for delivery of a fluid. In some embodiments, the system has one syringe. In some embodiments, the system has two syringes. In some embodiments, the system has three syringes. In some embodiments, the system has four or more syringes. The fluids suitable for use in the methods of the invention include those described herein, for example, one or more fluids each comprising an effective amount of one or more vectors as described herein, and one or more fluids comprising one or more therapeutic agents. id="p-114" id="p-114" id="p-114" id="p-114" id="p-114"
[0114] In some embodiments, the kit comprises a single fluid (e.g., a pharmaceutically acceptable fluid comprising an effective amount of the vector). In some embodiments, the kit comprises 2 fluids. In some embodiments, the kit comprises 3 fluids. In some embodiments, the kit comprises 4 or more fluids. A fluid may include a diluent, buffer, excipient, or any other liquid described herein or known in the art suitable for delivering, diluting, stabilizing, buffering, or otherwise transporting a rAAV vector composition of the present disclosure. In some embodiments, the kit comprises one or more buffers, e.g., an aqueous pH buffered solution. Examples of buffers may include without limitation phosphate, citrate, Tris, HEPES, and other organic acid buffers. id="p-115" id="p-115" id="p-115" id="p-115" id="p-115"
[0115] In some embodiments, the kit comprises a container. Suitable containers may include, e.g., vials, bags, syringes, and bottles. The container may be made of one or more of a material such as glass, metal, or plastic. In some embodiments, the container is used to hold a rAAV 259964/3 composition of the present disclosure. In some embodiments, the container may also hold a fluid and/or other therapeutic agent. id="p-116" id="p-116" id="p-116" id="p-116" id="p-116"
[0116] In some embodiments, the kit comprises an additional therapeutic agent with a rAAV composition of the present disclosure. In some embodiments, the rAAV composition and the additional therapeutic agent may be mixed. In some embodiments, the rAAV composition and the additional therapeutic agent may be kept separate. In some embodiments, the rAAV composition and the additional therapeutic agent may be in the same container. In some embodiments, the rAAV composition and the additional therapeutic agent may be in different containers. In some embodiments, the rAAV composition and the additional therapeutic agent may be administered simultaneously. In some embodiments, the rAAV composition and the additional therapeutic agent may be administered on the same day. In some embodiments, the rAAV composition may be administered within one day, two days, three days, four days, five days, six days, seven days, two weeks, three weeks, four weeks, two months, three months, four months, five months, or six months of administration of the additional therapeutic agent. id="p-117" id="p-117" id="p-117" id="p-117" id="p-117"
[0117] In some embodiments, the kit comprises a therapeutic agent to transiently suppress the immune system prior to AAV administration. In some embodiments, patients are transiently immune suppressed shortly before and after injection of the virus to inhibit the T cell response to the AAV particles (e.g. see Ferreira et al., Hum. Gene Ther. 25:180-188, 2014). In some embodiments, the kit further provides cyclosporine, mycophenolate mofetil, and/or methylprednisolone. id="p-118" id="p-118" id="p-118" id="p-118" id="p-118"
[0118] The rAAV particles and/or compositions of the invention may further be packaged into kits including instructions for use. In some embodiments, the kits further comprise a device for delivery (e.g., any type of parenteral administration described herein) of compositions of rAAV particles. In some embodiments, the instructions for use include instructions according to one of the methods described herein. In some embodiments, the instructions are printed on a label provided with (e.g., affixed to) a container. In some embodiments, the instructions for use include instructions for administering to a mammal (e.g., a human) an effective amount of rAAV particles, e.g., for treating mucolipidosis type II (ML II) and/or mucolipidosis type III (ML III), increasing body size, increasing bone mineral content, and/or increasing bone mineral density. 259964/3 VII. Animal Models id="p-119" id="p-119" id="p-119" id="p-119" id="p-119"
[0119] The invention provides animal models of Mucolipidosis II. GNPTAB mutant mice may be generated by microinjection of embryonic stem (ES) cell clones into host blastocysts using standard methods. Briefly, targeted disruption of the mouse GNPTAB locus (e.g., from exon 12 to exon 20 was deleted) may be performed by homologous recombination with a replacement vector containing a reporter or selectable marker gene. All offsprings were genotyped by polymerase chain reaction analysis in tail snip DNA. All mice used in this study were males identified as wild type (+/+) mice or heterozygous (+/-) or homozygous (-/-). In some embodiments, at least one allele of the GNPTAB gene comprises a deletion located between exons 12 and exon 20. In some embodiments, at least one allele of the GNPTAB gene comprises a deletion spanning exons 12 and exon 20. In some embodiments, the deletion comprises a deletion of one or more of exons 12, 13, 14, 15, 16, 17, 18, 19 or 20. In some embodiments, a portion of the GNPTAB gene is replaced by a gene encoding a reporter and/or selectable marker. In some embodiments, the selectable marker confers resistance to neomycin. In some embodiments, the animal is a mammal (e.g., a rodent, a rabbit, a cat, a dog, a pig, a monkey). In some embodiments, the mammal is a rodent (e.g., a mouse, a rat, a hamster, a guinea pig). In some embodiments, the animal is immunocompetent or immunodeficient. In some embodiments, the animal model comprises a genetically modified animal. Other mouse models of ML II are provided by Gelfman etal., (Invest. Optham. VisualSci. 2007, 48:5221­5228) and Paton, L. et al, (JBiol Chem. 2014, 289(39):26709-21). id="p-120" id="p-120" id="p-120" id="p-120" id="p-120"
[0120] In some aspects, the invention provides a method for evaluating an agent for treatment of Mucolipidosis II (ML II) comprising administering the agent to the animal model as described herein, wherein amelioration one or more symptoms of ML II indicates the agent may provide beneficial treatment of ML II. In some embodiments, the symptom of ML II is decreased body weight, decreased bone density, decreased bone mineral content, skeletal defects, cognitive deficits, delays in the development of gross and fine motor skills, hearing loss, lack of muscle tone, protruded abdomen, umbilical hernias, progressive mucosal thickening of the airways, frequent respiratory infections, thickening and insufficiency of the mitral valve, constipation and/or diarrhea. In some embodiments, the agent is a small molecule, a polypeptide, an antibody, a nucleic acid or a recombinant viral particle. 259964/3 EXAMPLES id="p-121" id="p-121" id="p-121" id="p-121" id="p-121"
[0121] The invention will be more fully understood by reference to the following examples. They should not, however, be construed as limiting the scope of the invention. It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims.
Example 1: Generation and characterization of GNPTAB knockout mice id="p-122" id="p-122" id="p-122" id="p-122" id="p-122"
[0122] Mucolipidosis type II (ML type II or ML-II, aka I-cell disease; OMIM entry #252500) is an autosomal recessive lysosomal storage disorder caused by N-acetylglucosamine- 1-phosphotransferase (GNPTAB) deficiency. The presence of numerous inclusion bodies in the cytoplasm of fibroblasts, a lack of mucopolysacchariduria, increased lysosomal enzyme activities in serum, and decreased GlcNAc-phosphotransferase activity are hallmarks of this disease. In order to study the pathology of ML type II, a GNPTAB knockout (KO) mouse was developed and characterized.
Methods Generation of AAV2/8-GNPTAB constructs[0123] The coding sequence of mouse GNPTAB was amplified and codon-optimized for expression in mice by Genescript (Piscataway, NJ, USA). Because of the large size of the GNPTAB cDNA and the limited capacity of AAV, the cDNA does not fit into any available vectors. Therefore a new expression cassette was designed that contains shortened versions of the CMV enhancer and chicken beta-actin promoter, as well as a very small intron. Expression of full-length GNPTAB mRNAwas confirmed by transient infection of HEK293 cells in vitro followed by quantitative RT-PCR. This AAV2/8-GNPTAB was purified, stored at -70°C, and used at a concentration of 2.5x1012 DNase resistant particles/mL. The shortened enhancer and promoter sequences were used for these studies. 259964/3 Animals[0124] GNPTAB mutant mice were generated by microinjection of embryonic stem (ES) cell clones into host blastocysts using standard methods. Briefly, targeted disruption of the mouse GNPTAB locus (from exon 12 to exon 20 was deleted) was performed by homologous recombination with a replacement vector containing the neomycin-resistance gene. Mice used in this study were of mixed genetic background (129/Sv and C57BL/6). All offsprings were genotyped by polymerase chain reaction analysis in tail snip DNA. All mice used in this study were males identified as wild type (+/+) mice or heterozygous (+/-) or homozygous (-/-). id="p-125" id="p-125" id="p-125" id="p-125" id="p-125"
[0125] Mice were housed in groups of 2-5 per cage in a colony room under a 12-hour light- dark cycle. Rodent diet (Harlan Teklad #8604, Madison, WI, USA) and water were available ad libitum. Care of animals was conducted in accordance with the guidelines described in the Guide for the Care and Use of Laboratory Animals (National Academy Press, Washington DC, 1996).
Vector injections[0126] A single bilateral i.v. injection of PBS or AAV-GNPTAB was made into 6-week-old GNPTAB KO mice. A group of wild type or heterozygous mice injected with PBS served as a control group. Each mouse injected with AAV-GNPTAB received 3x10״ vector particles. To reduce or eliminate immune response, anti-CD40 ligand antibody (MR1) alone was injected according to protocol by Halbert el al., (1998) J. Virol. 72:9795-9805.
Auxology[0127] All mice were examined weekly by measuring total body weight (g) and body length (nose-to-anus distance, mm), using an electronic digital caliper.
Histology[0128] Wild type and GNPTAB-/- mice at 10 mo. of age were sedated and perfused via the heart with PBS. Following the perfusion, the femurs were removed, fixed in 4% paraformaldehyde (PFA), decalcified with 0.5 M EDTA, and paraffin embedded. Femur sections (4 ^M) were stained with hematoxlin-eosin. Aperio ScanScope AT (v101.0) was used for scanning of slide and analysis at 4°C. The tissues were embedded and cut for TEM or hematoxylin and eosin (H&E). 259964/3 Ultrastructural analysis[0129] Wild type and KO mice were decapitated, and the salivary glands were removed and fixed by immersion in 2.5% glutaraldehyde in PBS overnight at 4°C. The salivary glands were dehydrated and then washed in PBS buffer for a further 30 min followed by post-fixation with 1% osmium tetroxide for 1 h at room temperature. The specimens were dehydrated through graded alcohol series and embedded in Epon 812. Semi-thin serial sections were cut at 2.(Leica ultracut UCT), stained with Toluidine Blue and observed under a light microscope. Transmission electron microscopy observation was performed with a Mirgagni microscope (FEI).
Lysosomal enzyme assay[0130] Blood was taken and samples were centrifuged 15 min (8000xg) and plasma was stored at -70°C. Plasma lysosomal enzyme activities were measured at the Department of Chemical Pathology, Samsung Medical Center, South Korea. In ML II, specific GlcNAc- phosphotransferase activity was not able to be measured. Thus, a diagnosis was made based on elevated plasma lysosomal enzyme.
DEXA analysis[0131] Bone mineral density (BMD), bone mineral content (BMC), and lean mass were measured in anesthetized mice with the pDEXA Sabre X-ray Bone Densitometer. After anesthesia was administered, the weight of each mouse was recorded, and the mouse was then placed in the DEXA scanner. For data analysis, a region comprising the entire body of the mouse was defined.
Real-time PCR[0132] Real-time polymerase chain reaction is performed to quantify the mRNA levels of GNPTAB using an ABI PRISM 7900HT system and TaqMan gene expression assays (Applied Biosystems, Foster City, CA). The mRNA levels are expressed relative to the level of GAPDH. The 2-AACT method is used to analyze the data using SDS2.3 software (Applied Biosystems). 259964/3 Statistical Analysis[0133] GraphPad Prism Version 5 was used for statistical analysis and drawing figures and tables. Significance of differences was determined by an unpaired Student’s /-test, and one-way ANOVA test. Unless otherwise specified, all data are presented as mean±SEM.
Results id="p-134" id="p-134" id="p-134" id="p-134" id="p-134"
[0134] A gene trap strategy was used to generate GNPTAB KO mice. As illustrated in FIG. 1A, homologous recombination with a replacement vector containing the neomycin-resistance gene between exon 12 and 20 was used to generate the mice. As shown in FIG. 1B, the presence of the gene trap in the GNPTAB gene of the ES clones was confirmed by Southern blot analysis. id="p-135" id="p-135" id="p-135" id="p-135" id="p-135"
[0135] A comprehensive phenotypic analysis was performed on wild type, heterozygous, and homozygous animals. KO mice could be visually distinguished from wild-type (WT) littermates by their small size. Mean body weight (FIG. 2A) and length (FIG. 2B) were significantly reduced in the KO mice. Consistent with their small size, the homozygous mice exhibited a coarse facial shape (FIG. 2C). id="p-136" id="p-136" id="p-136" id="p-136" id="p-136"
[0136] As shown in FIG. 3A, in normal cartilage, clear lacunar space surrounded chondrocytes. The cytoplasm contained a single, clear vacuole. In KO mice, femoral chondrocytes were markedly hypertrophic and completely filled their enlarged lacunae (FIG.3B). The cytoplasm of hypertrophic chondrocyte was distended by abundant microvacuoles containing scant amounts of finely granular amphophilic material. There was less fixation-related shrinkage of chondrocytes, which filled the enlarged lacunae. This contrasts with wild type chondrocytes, which contained a single large, clear vacuole and only partially filled lacunae. id="p-137" id="p-137" id="p-137" id="p-137" id="p-137"
[0137] The acini of the exocrine salivary glands, which consist of mucous- and serous-type secretory cells, exhibited extensive vacuolization in KO mice when examined by light microscopy (Gelfman etal., 2007, Invest. Optham. VisualSci. 48:5221-5228). To gain insight into the underlying pathology, the submandibular salivary gland was analyzed by electron microscopy (EM). id="p-138" id="p-138" id="p-138" id="p-138" id="p-138"
[0138] Both mucous- and serous-type secretory cells were readily observed in the wild type mice (FIG. 4A-C). By contrast, the overall architecture of the submandibular salivary gland of 259964/3 the KO mice was so grossly disrupted that it greatly hampered the identification of the serous cells in the EM sections (FIG. 4D-F). The mucous-type secretory cells were packed with large membrane-bound vacuoles that contained heterogeneous material (FIG. 4E, F). The large vacuoles were filled with un-degraded cytoplasmic material that accumulated in the KO mucous cells and were surrounded by a single membrane. These observations indicate that they likely represent autolysosomes formed by fusion of autophagic compartments with lysosomes. id="p-139" id="p-139" id="p-139" id="p-139" id="p-139"
[0139] Patients with ML-II have greatly elevated levels of lysosomal enzymes in their sera because of the inability to synthesize the mannose-6-phosphate recognition marker that is essential for proper targeting of these enzymes to lysosomes. This trafficking defect results in hypersecretion of the enzymes into the blood. FIGs. 5A-5D shows that compared with wild type mice, the KO mice exhibited great increased levels of lysosomal enzymes, such as N- acetylglucosaminidase (FIG. 5A), P-hexosaminidase A (FIG. 5B), P-galactosidase (FIG. 5C), and P-glucuronidase (FIG. 5D). This phenotype would be expected if GlcNAc-1- phosphotransferase activity were defective in the homozygous mice, consistent with observations in humans. id="p-140" id="p-140" id="p-140" id="p-140" id="p-140"
[0140] In summary, these experiments demonstrate that GNPTAB KO mice displayed clear phenotypic differences as compared to wild-type mice, particularly with respect to growth, salivary gland morphology, and lysosomal enzyme levels. These results indicate that GNPTAB KO mice represent a model system in which to study therapeutic treatments for ML-II.
Example 2: Evaluation of AAV-mediated administration of GNPTAB in GNPTAB KO mice id="p-141" id="p-141" id="p-141" id="p-141" id="p-141"
[0141] Because patients with ML-II show growth retardation, a major goal of this study was to evaluate the efficacy for AAV-mediated GNPTAB administration that may promote growth in an ML-II model. Therefore, the phenotypes of wild-type, GNPTAB heterozygous, and GNPTAB KO mice were analyzed, as compared to GNPTAB KO mice injected with AAV-GNPTAB. id="p-142" id="p-142" id="p-142" id="p-142" id="p-142"
[0142] As shown in FIGs. 6A and 6B, all analytical assessments were carried out during two periods post-injection: the first began 16 weeks post-injection (on 12 weeks old age), the second 259964/3 began 32 weeks post-injection (on 38 weeks old age). Auxological analysis was added 6 weeks post-injection. id="p-143" id="p-143" id="p-143" id="p-143" id="p-143"
[0143] A plasmid was constructed that contained an expression cassette expressing the mouse GNPTAB cDNA regulated by the CMV enhancer/CBA promoter and a BGH polyA signal (FIG. 7A). Full length GNPTAB was under the control of the shortened CMV enhance/CBA promoter as described herein. Quantitative real-time PCR analyses of livers from GNPTAB KO mice injected with AAV2/8-GNPTAB indicated specific and highly expression of AAV-GNPTAB. As expected, any AAV-GNPTAB mRNA was absent in samples from control littermates (FIG. 7B). id="p-144" id="p-144" id="p-144" id="p-144" id="p-144"
[0144] As an initial test to determine whether AAV-mediated gene transfer affects metabolism in a physiologically relevant way, weight gain was monitored for 32 weeks post injection. As shown in FIG. 8A, no effect was observed for treatment of AAV-GNPTAB in KO mice. FIG.8B shows that there was no difference observed between control and AAV-GNPTAB treated KO mice in terms of weight gained. id="p-145" id="p-145" id="p-145" id="p-145" id="p-145"
[0145] Next, the effectiveness of attenuating the dwarfism phenotype of KO mice was assessed. An improvement in dwarfism was noted after 6 weeks of therapy in KO mice injected AAV-GNPTAB, showing an increase in body size (FIGS. 9A, 9B and Table 1 below).
Table 1. Height change Genotype Starting height (mm) Ratio of height increase (Height after indicated interval/starting height)Height after 6 weeks Height after 32 weeks Wild-type (+/+) 84.31±0.885 1.06±0.011 1.11±0.008Heterozygous (+/-) 84.83±0.475 1.07±0.008 1.11±0.005GNPTAB knockout (-/- )GNPTAB knockout (-/- ) + AAV-GNPTAB 76.09±2.483 1.04±0.012 1.09±0.015 75.10±1.964 1.07±0.009 1.11±0.017Values depicted are mean ± SEM. id="p-146" id="p-146" id="p-146" id="p-146" id="p-146"
[0146] Next, bone mineral density (BMD), bone mineral content (BMC), and body composition were measured. DEXA is an x-ray-based imaging technique for determination of bone mineral and body composition (as body fat mass). AAV-GNPTAB transfer induced relative 259964/3 increases in BMC and BMD in AAV injected KO mice. FIGs. 10A-10C shows the raw BMD data obtained before injection (FIG. 10A) and relative ratio in BMD (FIGS. 10B, 10C) compared before and post injection. These data are also provided in Table 2 below. These results demonstrate a significant effect of the gene transfer on bone mineral density.
Table 2. Bone mineral density (BMD) change Genotype Starting BMD (g/cm2) Ratio of height increase (Height after indicated interval/starting height)BMD after 16 weeks BMD after 32 weeks Wild-type (+/+) 0.055±0.001 1.139±0.017 1.124±0.032Heterozygous (+/-) 0.056±0.001 1.137±0.016 1.140±0.007GNPTAB knockout (-/- )GNPTAB knockout (-/- ) + AAV-GNPTAB 0.050±0.002 1.156±0.011 1.157±0.012 0.048±0.001 1.219±0.013 1.236±0.044Values depicted are mean ± SEM. id="p-147" id="p-147" id="p-147" id="p-147" id="p-147"
[0147] Similarly, the raw bone mineral content (BMC) data showed a strong gene therapy effect (FIG. 11A). The ratio data (FIG. 11B, 11C) also approached a significant effect in bone growth. These data are also provided in Table 3 below.
Table 3. Bone mineral content (BMC) change Genotype Starting BMD (g/cm2) Ratio of height increase (Height after indicated interval/starting height)BMD after 16 weeks BMD after 32 weeks Wild-type (+/+) 0.575±0.016 1.172±0.047 1.123±0.073Heterozygous (+/-) 0.565±0.018 1.197±0.045 1.116±0.042GNPTAB knockout (-/- )GNPTAB knockout (-/- ) + AAV-GNPTAB 0.448±0.027 1.272±0.044 1.228±0.049 0.386±0.016 1.560±0.080 1.551±0.096Values depicted are mean ± SEM. id="p-148" id="p-148" id="p-148" id="p-148" id="p-148"
[0148] Next, analysis of body lean content using a DEXA scanner revealed that the lean mass was also reduced in the KO mice (FIG. 12A). As shown in FIG. 12B, 32 weeks after injection, control mice showed a significant decrease in the percentage of lean mass in comparison to data 259964/3 from before injection. However, no significant changes in lean mass were observed in AAV- GNPTAB treated KO mice. These data are also provided in Table 4 below.
Table 4. Change in percent lean mass Genotype Starting % Lean Mass% Lean Mass after weeksWild-type (+/+) 86.56±0.627 78.98±3.425Heterozygous (+/-) 87.44±0.321 69.5±3.429GNPTAB knockout (-/-) 85.86±0.548 70.2±3.313GNPTAB knockout (-/-)+ AAV-GNPTAB84.3±0.665 84.7±0.7087Values depicted are mean ± SEM. id="p-149" id="p-149" id="p-149" id="p-149" id="p-149"
[0149] In summary, GNPTAB KO mice failed to thrive and had weak bone density, as is the case in human ML type II. Using this model, the potential of gene therapy using an AAV vector for treatment of ML type II was examined. Overexpressed GNPTAB was found to partially protect bone growth impairment in KO mice. Altogether, the systemic delivery of GNPTAB through AAV vectors was highly efficient and represents a promising approach for the correction of the bone pathology in ML type II. 259964/3 SEQUENCES All polypeptide sequences are presented N-terminal to C-terminal unless otherwise noted. All nucleic sequences are presented 5’ to 3’ unless otherwise noted.
GNPTAB Human GNPTAB Protein Sequence (SEQ ID NO:1) MLFKLLQRQTYTCLSHRYGLYVCFLGVVVTIVSAFQFGEVVLEWSRDQYHVLFDSYRDNIAGKSFQNRLC LPMPIDVVYTWVNGTDLELLKELQQVREQMEEEQKAMREILGKNTTEPTKKSEKQLECLLTHCIKVPMLV LDPALPANITLKDLPSLYPSFHSASDIFNVAKPKNPSTNVSVVVFDSTKDVEDAHSGLLKGNSRQTVWRG YLTTDKEVPGLVLMQDLAFLSGFPPTFKETNQLKTKLPENLSSKVKLLQLYSEASVALLKLNNPKDFQEL NKQTKKNMTIDGKELTISPAYLLWDLSAISQSKQDEDISASRFEDNEELRYSLRSIERHAPWVRNIFIVT NGQIPSWLNLDNPRVTIVTHQDVFRNLSHLPTFSSPAIESHIHRIEGLSQKFIYLNDDVMFGKDVWPDDF YSHSKGQKVYLTWPVPNCAEGCPGSWIKDGYCDKACNNSACDWDGGDCSGNSGGSRYIAGGGGTGSIGVG QPWQFGGGINSVSYCNQGCANSWLADKFCDQACNVLSCGFDAGDCGQDHFHELYKVILLPNQTHYIIPKG ECLPYFSFAEVAKRGVEGAYSDNPIIRHASIANKWKTIHLIMHSGMNATTIHFNLTFQNTNDEEFKMQIT VEVDTREGPKLNSTAQKGYENLVSPITLLPEAEILFEDIPKEKRFPKFKRHDVNSTRRAQEEVKIPLVNI SLLPKDAQLSLNTLDLQLEHGDITLKGYNLSKSALLRSFLMNSQHAKIKNQAIITDETNDSLVAPQEKQV HKSILPNSLGVSERLQRLTFPAVSVKVNGHDQGQNPPLDLETTARFRVETHTQKTIGGNVTKEKPPSLIV PLESQMTKEKKITGKEKENSRMEENAENHIGVTEVLLGRKLQHYTDSYLGFLPWEKKKYFQDLLDEEESL KTQLAYFTDSKNTGRQLKDTFADSLRYVNKILNSKFGFTSRKVPAHMPHMIDRIVMQELQDMFPEEFDKT SFHKVRHSEDMQFAFSYFYYLMSAVQPLNISQVFDEVDTDQSGVLSDREIRTLATRIHELPLSLQDLTGL EHMLINCSKMLPADITQLNNIPPTQESYYDPNLPPVTKSLVTNCKPVTDKIHKAYKDKNKYRFEIMGEEE IAFKMIRTNVSHVVGQLDDIRKNPRKFVCLNDNIDHNHKDAQTVKAVLRDFYESMFPIPSQFELPREYRN RFLHMHELQEWRAYRDKLKFWTHCVLATLIMFTIFSFFAEQLIALKRKIFPRRRIHKEASPNRIRV Mouse GNPTAB Protein Sequence (SEQ ID NO:2) MLLKLLQRQTYTCLSHRYGLYVCFVGVVVTIVSAFQFGEVVLEWSRDQYHVLFDSYRDNIAGKSFQNRLC LPMPIDVVYTWVNGTDLELLKELQQVREHMEEEQRAMRETLGKNTTEPTKKSEKQLECLLTHCIKVPMLV LDPPLPANCTLKDLPTLYPSFHAASDMFNVAKPKNPSTNVSVVVFDTTKDVEDAHAGPFKGGSKQMVWRA YLTTDKEAPGLVLMQGLAFLSGFPPTFKETSQLKTKLPEKLSSKIKLLRLYSEASVALLKLNNPKGFQEL NKQTKKNMTIDGKELTISPAYLLWDLSAISQSKQDEDVSASRFEDNEELRYSLRSIERHAPWVRNIFIVT NGQIPSWLNLDNPRVTIVTHQDIFQNLSHLPTFSSPAIESHIHRIEGLSQKFIYLNDDVMFGKDVWPDDF YSHSKGQKVYLTWPVPNCAEGCPGSWIKDGYCDKACNNSACDWDGGDCSGNTAGNRFVAGGGGTGNIGAG QHWQFGGGINTISYCNQGCANSWLADKFCDQACNVLSCGFDAGDCGQDHFHELYKVTLLPNQTHYVVPKG EYLSYFSFANIARRGVEGTYSDNPIIRHASIANKWKTIHLIMHSGMNATTIYFNLTLQNANDEEFKIQIA VEVDTREAPKLNSTTQKAYESLVSPVTPLPQADVPFEDVPKEKRFPKIRRHDVNATGRFQEEVKIPRVNI SLLPKEAQVRLSNLDLQLERGDITLKGYNLSKSALLRSFLGNSLDTKIKPQARTDETKGNLEVPQENPSH RRPHGFAGEHRSERWTAPAETVTVKGRDHALNPPPVLETNARLAQPTLGVTVSKENLSPLIVPPESHLPK EEESDRAEGNAVPVKELVPGRRLQQNYPGFLPWEKKKYFQDLLDEEESLKTQLAYFTDSKHTGRQLKDTF ADSLRYVNKILNSKFGFTSRKVPAHMPHMIDRIVMQELQDMFPEEFDKTSFHKVRHSEDMQFAFSYFYYL MSAVQPLNISQVFHEVDTDQSGVLSDREIRTLATRIHDLPLSLQDLTGLEHMLINCSKMLPANITQLNNI PPTQEAYYDPNLPPVTKSLVTNCKPVTDKIHKAYKDKNKYRFEIMGEEEIAFKMIRTNVSHVVGQLDDIR KNPRKFVCLNDNIDHNHKDARTVKAVLRDFYESMFPIPSQFELPREYRNRFLHMHELQEWRAYRDKLKFW THCVLATLIIFTIFSFFAEQIIALKRKIFPRRRIHKEASPDRIRV

Claims (25)

1./ - 51 - CLAIMS 1. A recombinant adeno-associated virus (rAAV) vector comprising nucleic acid encoding N-acetylglucosamine-1-phosphate transferase (GNPTAB) and at least one AAV inverted terminal repeat (ITR), wherein the vector comprises a CMV enhancer/chicken beta-actin (CBA) promoter, wherein: (i) the CBA promoter is a modified CBA promoter, optionally a truncated CBA promoter, and/or (ii) the CMV enhancer is a shortened CMV enhancer.
2. The rAAV vector of claim 1, wherein: (a) the GNPTAB comprises the alpha and beta subunits; (b) the GNPTAB is operably linked to a promoter; (c) the GNPTAB is a human GNPTAB; (d) the GNPTAB comprises an amino acid sequence that is at least about 80%, at least about 85%, at least about 90% or at least about 95% identical to the amino acid sequence of SEQ ID NO:1; (e) the GNPTAB comprises the amino acid sequence of SEQ ID NO:1; and/or (f) the AAV terminal repeat is an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAVrh8R, AAV9, AAV10, AAVrh10, AAV11, AAV12, AAV2R471A, AAV DJ, a goat AAV, bovine AAV, or mouse AAV serotype ITR.
3. The rAAV vector of claim 1 or 2, wherein the vector comprises: (a) an intron; (b) a polyadenylation sequence; and/or (c) two ITRs.
4. The rAAV vector of any one of claims 1-3, wherein the vector comprises: (a) an intron, wherein the intron is an MVM intron; (b) a polyadenylation sequence, wherein the polyadenylation sequence is a bovine growth hormone polyadenylation sequence; and/or 259964/ - 52 - (c) two ITRs.
5. A rAAV particle comprising the rAAV vector of any one of claims 1-4, wherein the rAAV particle comprises: an AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAVrh8R, AAV9, AAV10, AAVrh10, AAV11, AAV12, AAV2R471A, AAV2/2-7m8, AAV DJ, AAV2 N587A, AAV2 E548A, AAV2 N708A, AAV V708K, goat AAV, AAV1/AAVchimeric, bovine AAV, mouse AAV, or rAAV2/HBoV1 serotype capsid.
6. An rAAV particle comprising the rAAV vector of any one of claims 1-4, wherein the at least one ITR and the capsid are derived from the same AAV serotype, or the at least one ITR is derived from a different AAV serotype than the capsid of the rAAV viral particle.
7. The rAAV particle of claim 5 or 6, wherein the rAAV particle comprises an AAVcapsid and AAV2 ITRs.
8. The rAAV particle of any one of claims 5-7, wherein the rAAV particle is produced by transfecting a host cell with nucleic acid encoding the rAAV vector and nucleic acid encoding AAV rep and cap functions, and providing nucleic acid encoding AAV helper functions, wherein the AAV helper functions are provided by transfecting the host cell with nucleic acid encoding the AAV helper functions or the AAV helper functions are provided by infecting the host cell with an AAV helper virus that provides the AAV helper functions, optionally the AAV helper virus is an adenovirus, a herpes simplex virus or a baculovirus.
9. The rAAV particle of any one of claims 5-7, wherein the rAAV particle is produced by an AAV producer cell comprising nucleic acid encoding the rAAV vector and nucleic acid encoding AAV rep and cap functions, and providing nucleic acid encoding AAV helper functions.
10. A pharmaceutical composition comprising the rAAV particle of any one of claims 5-9. 259964/ - 53 -
11. A rAAV particle of any one of claims 5-9, or the pharmaceutical composition of claim for use in treating mucolipidosis type II (ML II) or mucolipidosis type III (ML III) in a mammal.
12. The rAAV particle or pharmaceutical composition of claim 11, wherein the treatment ameliorates or delays progression of one or more symptoms of ML II or ML III, wherein the one or more symptoms of ML II or ML III are skeletal defects, cognitive deficits, delays in the development of gross and fine motor skills, hearing loss, lack of muscle tone, protruded abdomen, umbilical hernias, progressive mucosal thickening of the airways, frequent respiratory infections, thickening and insufficiency of the mitral valve, constipation or diarrhea.
13. A rAAV particle comprising a rAAV vector, for use in maintaining, preventing the reduction of, or increasing body size, bone mineral content, or bone mineral density in a mammal with mucolipidosis type II (ML II) or mucolipidosis type III (ML III), wherein the rAAV vector comprises nucleic acid encoding N-acetylglucosamine-1-phosphate transferase (GNPTAB) and at least one AAV ITR, wherein expression of the GNPTAB results in maintenance of, prevention of the reduction of, or an increase in body weight; maintenance of or an increase in body height; and/or maintenance, prevention of reduction, or an increase in bone mineral content or density.
14. The rAAV particle of any one of claims 11-13, wherein the mammal is a human.
15. The rAAV particle of any one of claims 11-14, wherein the mammal is a pediatric subject or a young adult.
16. The rAAV particle of any one of claims 11-15, wherein: (a) the rAAV particle is administered intravenously, intraperitoneally, intra-arterially, intramuscularly, subcutaneously, or intrahepatically; (b) the rAAV particle is administered to more than one location; 259964/ - 54 - (c) the administration is repeated; and/or (d) the rAAV viral particles are in a pharmaceutical composition.
17. A rAAV particle of any one of claims 5-9 or a pharmaceutical composition of claim 10 in the manufacture of a medicament for use in treating ML II or ML III in a mammal, optionally a human, optionally the rAAV particle of any one of claims 11-16.
18. A rAAV particle of any one of claims 5-9 or a pharmaceutical composition of claim for use in the manufacture of a medicament for ameliorating one or more symptoms of ML II or ML III in a mammal, optionally a human, or delaying the progression of one or more symptoms of ML II or ML III in a mammal, optionally a human, optionally wherein the one or more symptoms of ML II or ML III are skeletal defects, cognitive deficits, delays in the development of gross and fine motor skills, hearing loss, lack of muscle tone, protruded abdomen, umbilical hernias, progressive mucosal thickening of the airways, frequent respiratory infections, thickening and insufficiency of the mitral valve, constipation or diarrhea.
19. A rAAV particle of any one of claims 5-9 or a pharmaceutical composition of claim for use for ameliorating one or more symptoms of ML II or ML III in a mammal, optionally a human, or delaying the progression of one or more symptoms of ML II or ML III in a mammal, optionally a human, optionally wherein the one or more symptoms of ML II or ML III are skeletal defects, cognitive deficits, delays in the development of gross and fine motor skills, hearing loss, lack of muscle tone, protruded abdomen, umbilical hernias, progressive mucosal thickening of the airways, frequent respiratory infections, thickening and insufficiency of the mitral valve, constipation or diarrhea.
20. A kit comprising the rAAV vector of any one of claims 1-4, the rAAV particle of any one of claims 5-9, or the pharmaceutical composition of claim 10.
21. The kit of claim 20, further comprising one or more buffers or pharmaceutically acceptable excipients and/or instructions for use in treating ML II and/or ML III. 259964/ - 55 -
22. A kit for treating ML II or ML III according to the method of any one of claims 11-16, wherein the kit comprises the rAAV vector of any one of claims 1-4, the rAAV particle of any one of claims 5-9, or the pharmaceutical composition of claim 10.
23. A method of generating an animal model of Mucolipidosis II (ML II) comprising introducing a deletion between exons 12 and 20 in at least one allele of the GNPTAB gene on the animal, optionally wherein at least one allele of the GNPTAB gene comprises a deletion spanning exons 12 and exon 20 and/or a portion of the GNPTAB gene is replaced by a gene encoding a reporter and/or selectable marker, optionally the selectable marker confers resistance to neomycin, further optionally wherein the animal is: (a) bred to be homozygous for the deletion in the GNPTAB gene; (b) bred to be heterozygous for the deletion in the GNPTAB gene; and/or (c) a mammal, optionally a rodent, optionally a mouse, further optionally the mouse has a genetic background derived from 129/Sv and/or C57Bl/6.
24. An animal model of Mucoliposis II (ML II), optionally an animal model generated by the method of claim 23, wherein at least one allele of a N-acetylglucosamine-1-phosphate transferase (GNPTAB) gene comprises a deletion located between exons 12 and exon 20, optionally at least one allele of the GNPTAB gene comprises a deletion spanning exons and exon 20 and/or a portion of the GNPTAB gene is replaced by a gene encoding a reporter and/or selectable marker, optionally the selectable marker confers resistance to neomycin, further optionally wherein the animal is: (a) homozygous or heterozygous for the deletion in the GNPTAB gene; and/or (b) a mammal, optionally a rodent, optionally a mouse, further optionally the mouse has a genetic background derived from 129/Sv and/or C57Bl/6.
25. A method for evaluating an agent for treatment of Mucolipidosis II (ML II) comprising administering the agent, optionally a small molecule, a polypeptide, an antibody, a nucleic acid or a recombinant viral particle, to the animal model of claim 24, wherein amelioration one or more symptoms of ML II indicates the agent may provide beneficial 259964/ - 56 - treatment of ML II, optionally wherein the symptom of ML II is decreased body weight, decreased bone density, decreased bone mineral content, skeletal defects, cognitive deficits, delays in the development of gross and fine motor skills, hearing loss, lack of muscle tone, protruded abdomen, umbilical hernias, progressive mucosal thickening of the airways, frequent respiratory infections, thickening and insufficiency of the mitral valve, constipation and/or diarrhea.
IL259964A 2015-12-15 2016-12-14 Adeno-associated viral vectors for treating mucolipidosis type ii IL259964B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562267502P 2015-12-15 2015-12-15
PCT/US2016/066611 WO2017106313A1 (en) 2015-12-15 2016-12-14 Adeno-associated viral vectors for treating mucolipidosis type ii

Publications (2)

Publication Number Publication Date
IL259964A IL259964A (en) 2018-07-31
IL259964B true IL259964B (en) 2022-09-01

Family

ID=57737987

Family Applications (1)

Application Number Title Priority Date Filing Date
IL259964A IL259964B (en) 2015-12-15 2016-12-14 Adeno-associated viral vectors for treating mucolipidosis type ii

Country Status (13)

Country Link
US (1) US20200261598A1 (en)
EP (1) EP3390644A1 (en)
JP (2) JP7347932B2 (en)
KR (1) KR20180098294A (en)
CN (1) CN108603199A (en)
AU (2) AU2016372035B2 (en)
BR (1) BR112018012015A2 (en)
CA (1) CA3008268A1 (en)
IL (1) IL259964B (en)
MX (1) MX2018007330A (en)
RU (1) RU2742612C2 (en)
SG (2) SG11201804994QA (en)
WO (1) WO2017106313A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2020118342A (en) 2017-11-07 2021-12-08 Дзе Юниверсити Оф Норт Каролина Эт Чепел Хилл OPTIMIZED AGA GENES AND EXPRESSION CLUSTERS AND THEIR APPLICATION
JP2022508182A (en) * 2018-11-21 2022-01-19 ストライドバイオ,インコーポレイテッド Recombinant viral vector and nucleic acid for its production

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008154198A1 (en) * 2007-06-06 2008-12-18 Genzyme Corporation Gene therapy for lysosomal storage diseases
WO2015168666A2 (en) * 2014-05-02 2015-11-05 Genzyme Corporation Aav vectors for retinal and cns gene therapy

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6989264B2 (en) 1997-09-05 2006-01-24 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
CN1990865A (en) * 1998-04-27 2007-07-04 联邦高等教育系统坦普尔大学 Method of treating endotoxemia
US6534300B1 (en) * 1999-09-14 2003-03-18 Genzyme Glycobiology Research Institute, Inc. Methods for producing highly phosphorylated lysosomal hydrolases
US6723551B2 (en) 2001-11-09 2004-04-20 The United States Of America As Represented By The Department Of Health And Human Services Production of adeno-associated virus in insect cells
CN103555678B (en) 2001-11-13 2018-02-09 宾夕法尼亚大学托管会 The method for the novel sequences that detection and/or identification adeno-associated virus (AAV) sequence and separation are identified
CN100471957C (en) * 2002-04-30 2009-03-25 艾维亚医药/生物技术有限公司 Adenovirus vectors for immunotherapy
US7510872B2 (en) 2003-02-26 2009-03-31 Nationwide Children's Hospital Recombinant adeno-associated virus production
PT1625210E (en) 2003-05-21 2011-03-15 Genzyme Corp Methods for producing preparations of recombinant aav virions substantially free of empty capsids
WO2006119432A2 (en) 2005-04-29 2006-11-09 The Government Of The U.S.A., As Rep. By The Sec., Dept. Of Health & Human Services Isolation, cloning and characterization of new adeno-associated virus (aav) serotypes
EP2007795B1 (en) 2006-03-30 2016-11-16 The Board Of Trustees Of The Leland Stanford Junior University Aav capsid proteins
ES2693194T3 (en) 2009-06-16 2018-12-10 Genzyme Corporation Improved methods for the purification of recombinant AAV vectors
LT2588130T (en) * 2010-06-25 2016-12-12 Shire Human Genetic Therapies, Inc. Cns delivery of therapeutic agents
US8663624B2 (en) 2010-10-06 2014-03-04 The Regents Of The University Of California Adeno-associated virus virions with variant capsid and methods of use thereof
EP2675902B1 (en) 2011-02-17 2019-03-27 The Trustees Of The University Of Pennsylvania Compositions and methods for altering tissue specificity and improving aav9-mediated gene transfer
JP2016517278A (en) * 2013-03-15 2016-06-16 ザ・チルドレンズ・ホスピタル・オブ・フィラデルフィアThe Children’S Hospital Of Philadelphia Vectors comprising stuffer / filler polynucleotide sequences and methods of use thereof
WO2015060722A1 (en) * 2013-10-24 2015-04-30 Uniqure Ip B.V. Aav-5 pseudotyped vector for gene therapy for neurological diseases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008154198A1 (en) * 2007-06-06 2008-12-18 Genzyme Corporation Gene therapy for lysosomal storage diseases
WO2015168666A2 (en) * 2014-05-02 2015-11-05 Genzyme Corporation Aav vectors for retinal and cns gene therapy

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ARONOVICH, ELENA L., AND PERRY B. HACKETT., LYSOSOMAL STORAGE DISEASE: GENE THERAPY ON BOTH SIDES OF THE BLOOD–BRAIN BARRIER., 28 February 2015 (2015-02-28) *
DATABASE UNIPROT [ONLINE], RECNAME: FULL=N-ACETYLGLUCOSAMINE-1-PHOSPHOTRANSFERASE SUBUNITS ALPHA/BETA; EC=2.7.8.17 {EC0:00002691PUBMED: 19955174}; ALTNAME: FULL=GLCNAC-1-PHOSPHOTRANSFERASE SUBUNITS ALPHA/BETA; ALTNAME: FULL=STEALTH PROTEIN GNPTAB;ALTNAME: FULL=UDP-N-ACETYLGLUC, 7 March 2006 (2006-03-07) *
GRAY, STEVEN J., ET AL., OPTIMIZING PROMOTERS FOR RECOMBINANT ADENO-ASSOCIATED VIRUS-MEDIATED GENE EXPRESSION IN THE PERIPHERAL AND CENTRAL NERVOUS SYSTEM USING SELF-COMPLEMENTARY VECTORS., 8 April 2011 (2011-04-08) *
PATON, LEIGH, ET AL., A NOVEL MOUSE MODEL OF A PATIENT MUCOLIPIDOSIS II MUTATION RECAPITULATES DISEASE PATHOLOGY., 26 September 2014 (2014-09-26) *
VOGEL, P., ET AL., COMPARATIVE PATHOLOGY OF MURINE MUCOLIPIDOSIS TYPES II AND IIIC., 1 March 2009 (2009-03-01) *

Also Published As

Publication number Publication date
JP2019500034A (en) 2019-01-10
KR20180098294A (en) 2018-09-03
WO2017106313A1 (en) 2017-06-22
SG11201804994QA (en) 2018-07-30
AU2016372035A1 (en) 2018-08-02
MX2018007330A (en) 2019-03-14
US20200261598A1 (en) 2020-08-20
BR112018012015A2 (en) 2018-12-04
JP7347932B2 (en) 2023-09-20
RU2018124448A (en) 2020-01-17
SG10201912935WA (en) 2020-02-27
CA3008268A1 (en) 2017-06-22
RU2742612C2 (en) 2021-02-09
IL259964A (en) 2018-07-31
AU2023204116A1 (en) 2023-07-13
RU2018124448A3 (en) 2020-08-14
CN108603199A (en) 2018-09-28
JP2022046635A (en) 2022-03-23
EP3390644A1 (en) 2018-10-24
AU2016372035B2 (en) 2023-03-30

Similar Documents

Publication Publication Date Title
US20220395586A1 (en) Enhanced delivery of viral particles to the striatum and cortex
EP3233129B1 (en) Nucleic acid constructs and gene therapy vectors for use in the treatment of wilson&#39;s disease and other conditions
Wolf et al. Direct gene transfer to the CNS prevents emergence of neurologic disease in a murine model of mucopolysaccharidosis type I
IL294965A (en) Production of oversized adeno-associated vectors
IL284741B2 (en) Gene therapy for retinitis pigmentosa
JP2018535955A (en) Method of treating neurodegenerative diseases using gene therapy to delay the onset and progression of the disease while providing cognitive protection
US10648000B2 (en) rAAV vector compositions, methods for targeting vascular endothelial cells and use in treatment of type I diabetes
AU2023204116A1 (en) Adeno-associated viral vectors for treating mucolipidosis type II
US20210348135A1 (en) Generation of improved human pah for treatment of severe pku by liver-directed gene replacement therapy
JP2024056832A (en) Adeno-associated virus vectors for treating mucolipidosis type II
US20220112520A1 (en) Human pah expression cassette for treatment of pku by liver-directed gene replacement therapy
US20230398192A1 (en) Methods of treating metachromatic leukodystrophy
CN116648503A (en) Human PAH expression cassettes for treatment of PKU by liver-directed gene replacement therapy
JP2023545384A (en) Recombinant adeno-associated virus for central nervous system or muscle delivery
CN112980857A (en) Nucleotide composition for coding secretory wild type GAA protein, adeno-associated virus vector, and medicine and application thereof